1
|
Zhang T, Zhou W, Fan T, Yuan Y, Tang X, Zhang Q, Zou J, Li Y. Lactic acid metabolism: gynecological cancer's Achilles' heel. Discov Oncol 2025; 16:657. [PMID: 40314877 PMCID: PMC12048388 DOI: 10.1007/s12672-025-02364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
Lactic acid is significantly expressed in many cancers, including gynecological cancer, and has become a key regulator of the proliferation, development, metastasis and invasion of these cancers. In clinical and experimental studies, the level of lactic acid in gynecological cancer is closely related to metastasis and invasion, tumor recurrence and poor prognosis. Lactic acid can regulate the internal metabolic pathway of gynecological cancer cells and drive the autonomous role of non-cancer cells in gynecological cancer. In addition to being used as a source of energy metabolism by gynecological cancer cells, lactic acid can also be transported from cancer cells to neighboring cancer cells, stroma and vascular endothelial cells (ECs) to further guide metabolic reprogramming. Lactic acid is also involved in promoting inflammation and angiogenesis in gynecologic tumors. Therefore, we reviewed the mechanisms and recent advances in the production and transport of lactic acid in gynecological cancer. These advances and evidence suggest that targeted lactic acid metabolism is a promising cancer treatment.
Collapse
Affiliation(s)
- Ting Zhang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Wenchao Zhou
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Tingyu Fan
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yuwei Yuan
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Qunfeng Zhang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Juan Zou
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Yukun Li
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
2
|
Paul BM, Sundararajan VV, Raj FJ, Kannan G, Durairajan MB, Thangaraj P. In silico docking, ADMET profiling, and bio-accessibility experimentation on Breynia retusa phytocompounds and in vitro validation for anti-proliferative potencies against ovarian carcinoma. 3 Biotech 2025; 15:121. [PMID: 40225420 PMCID: PMC11981996 DOI: 10.1007/s13205-025-04276-8] [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: 01/18/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
This study aimed to assess the medicinal properties of Breynia retusa, a plant rich in phytocompounds predominantly used as an ethnomedicinal agent in Western Ghats, which appeared to be promising for therapeutic use, especially in the treatment of ovarian cancer. Herein, its cytotoxic potential on ovarian cancer cell lines SKOV-3, neurotoxicity, antioxidant activity, and molecular docking was determined to aid in explaining the mechanisms of interactions with proteins related to ovarian cancer. B . retusa methanolic extract demonstrated exuberant antioxidant activity, with 81.91% scavenging ability of DPPH radicals and efficient reduction of phosphomolybdenum (22.98 mg ascorbic acid equivalents antioxidant capacity/g extract). The extract proved to be an important anti-inflammatory agent through membrane stabilization inhibition of 83%. The cytotoxicity study against the SKOV-3 cell line indicated an IC50 value of 34.01 µg/mL and a very negligible neurotoxicity in SH-SY5Y cell lines. The GC-MS and HPLC profiling indicated many anticancer compounds in the extract such as secalciferol, methyl gallate, ricinoleic acid, gallic acid, and naringenin. The docking study showed significant interactions of secalciferol molecules with the key ovarian cancer proteins, which include IGF1 (-6.758 kcal/mol) and c-ERBB2 (-4.281 kcal/mol). Fatty acid derivatives and methyl gallate showed efficient dock scores (< -5.0 kcal/mol) with antioxidant (catalase and superoxide dismutase) enzymes and inflammatory cytokines (IL-6 and COX-1), respectively, as evidences of antioxidant and anti-inflammatory potentials. The bio-accessibility of phenolics and their antioxidant activity ranged above 90%, indicating the promising bioavailability of phytochemicals expected in vivo. Hence the current study emphasizes the anticancer potential of B. retusa phytocompounds that appeared to interact very strongly with ovarian cancer targets and confirms the dose-dependent cytotoxic and antioxidant activities of B. retusa methanolic extract. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04276-8.
Collapse
Affiliation(s)
- Benedict Mathews Paul
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Vetri Velavan Sundararajan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Francis Jegan Raj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Gowtham Kannan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Madhu Bala Durairajan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046 India
| |
Collapse
|
3
|
Mir IA, Mir HA, Mehraj U, Bhat MY, Mir MA, Dar TA, Hussain MU. Chloroquine sensitises hypoxic colorectal cancer cells to ROS-mediated cell death via structural disruption of pyruvate dehydrogenase kinase 1. Free Radic Biol Med 2025; 227:656-666. [PMID: 39657842 DOI: 10.1016/j.freeradbiomed.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Chloroquine (CQ), an autophagy antagonist, has been recently explored as a repurposable medicine for cancer; however the exact mechanism of its action is still not known. The present study investigated the effect of CQ on colorectal cancer cells to elucidate the underlying molecular mechanisms. We report for the first time that CQ suppresses hypoxia-induced growth and survival of HCT-116 cells by reducing glycolytic capacity and NAD+ production through inhibition of PDK1. Furthermore, in silico and in vitro studies revealed that CQ induces structural alteration in the PDK1 protein, leading to its destabilization and promotes its enhanced degradation by proteases. This degradation is in turn inhibited by the MG-132 protease inhibitor. Moreover, CQ-induced suppression of PDK1 results in mitochondrial damage through excessive production of ROS, as reflected by the reduction in mitochondrial membrane potential, which in turn triggers apoptosis through PARP cleavage and Caspase activation. These findings advocate CQ as a promising repurposable chemotherapeutic for colorectal cancer and a novel inhibitor of PDK1.
Collapse
Affiliation(s)
- Irfan Ahmad Mir
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Hilal Ahmad Mir
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India; Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Umar Mehraj
- Department of Pathology, Duke University, Durham, NC 27710, USA; Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, J&K, India
| | - Mohd Younus Bhat
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, 190006, J&K, India
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India
| | - Mahboob-Ul Hussain
- Department of Biotechnology, School of Biological Sciences, University of Kashmir, Srinagar, 190006, India.
| |
Collapse
|
4
|
Kangra K, Kakkar S, Mittal V, Kumar V, Aggarwal N, Chopra H, Malik T, Garg V. Incredible use of plant-derived bioactives as anticancer agents. RSC Adv 2025; 15:1721-1746. [PMID: 39835210 PMCID: PMC11744461 DOI: 10.1039/d4ra05089d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Cancer is a major global concern. Despite considerable advancements in cancer therapy and control, there are still large gaps and requirements for development. In recent years, various naturally occurring anticancer drugs have been derived from natural resources, such as alkaloids, glycosides, terpenes, terpenoids, flavones, and polyphenols. Plant-derived substances exhibit their anticancer potential through antiproliferative activity, cytotoxicity, apoptosis, angiogenesis and cell cycle arrest. Natural compounds can affect the molecular activity of cells through various signaling pathways, like the cell cycle pathway, STAT-3 pathway, PI3K/Akt, and Ras/MAP-kinase pathways. Capsaicin, ouabain, and lycopene show their anticancer potential through the STAT-3 pathway in breast, colorectal, pancreatic, lung, cervical, ovarian and colon cancers. Epigallocatechin gallate and emodin target the JNK protein in skin, breast, and lung cancers, while berberine, evodiamine, lycorine, and astragalin exhibit anticancer activity against breast, liver, prostate, pancreatic and skin cancers and leukemia through the PI3K/Akt and Ras/MAP-kinase pathways. In vitro/in vivo investigations revealed that secondary metabolites suppress cancer cells by causing DNA damage and activating apoptosis-inducing enzymes. After a meticulous literature review, the anti-cancer potential, mode of action, and clinical trials of 144 bioactive compounds and their synthetic analogues are included in the present work, which could pave the way for using plant-derived bioactives as anticancer agents.
Collapse
Affiliation(s)
- Kiran Kangra
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Saloni Kakkar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Virender Kumar
- College of Pharmacy, Pandit Bhagwat Dayal Sharma University of Health Sciences Rohtak 124001 India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana Ambala 133207 Haryana India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences Chennai 602105 Tamil Nadu India
| | - Tabarak Malik
- Department of Biomedical Sciences, Jimma University Jimma Ethiopia
- Division of Research & Development, Lovely Professional University Phagwara Punjab-144411 India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| |
Collapse
|
5
|
Kooshan Z, Cárdenas-Piedra L, Clements J, Batra J. Glycolysis, the sweet appetite of the tumor microenvironment. Cancer Lett 2024; 600:217156. [PMID: 39127341 DOI: 10.1016/j.canlet.2024.217156] [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: 02/14/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Cancer cells display an altered metabolic phenotype, characterised by increased glycolysis and lactate production, even in the presence of sufficient oxygen - a phenomenon known as the Warburg effect. This metabolic reprogramming is a crucial adaptation that enables cancer cells to meet their elevated energy and biosynthetic demands. Importantly, the tumor microenvironment plays a pivotal role in shaping and sustaining this metabolic shift in cancer cells. This review explores the intricate relationship between the tumor microenvironment and the Warburg effect, highlighting how communication within this niche regulates cancer cell metabolism and impacts tumor progression and therapeutic resistance. We discuss the potential of targeting the Warburg effect as a promising therapeutic strategy, with the aim of disrupting the metabolic advantage of cancer cells and enhancing our understanding of this complex interplay within the tumor microenvironment.
Collapse
Affiliation(s)
- Zeinab Kooshan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Lilibeth Cárdenas-Piedra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell & Tissue Engineering Technologies, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell & Tissue Engineering Technologies, Brisbane, Australia.
| |
Collapse
|
6
|
Şeker Karatoprak G, Dumlupınar B, Celep E, Kurt Celep I, Küpeli Akkol E, Sobarzo-Sánchez E. A comprehensive review on the potential of coumarin and related derivatives as multi-target therapeutic agents in the management of gynecological cancers. Front Pharmacol 2024; 15:1423480. [PMID: 39364049 PMCID: PMC11447453 DOI: 10.3389/fphar.2024.1423480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/16/2024] [Indexed: 10/05/2024] Open
Abstract
Current treatments for gynecological cancers include surgery, radiotherapy, and chemotherapy. However, these treatments often have significant side effects. Phytochemicals, natural compounds derived from plants, offer promising anticancer properties. Coumarins, a class of benzopyrone compounds found in various plants like tonka beans, exhibit notable antitumor effects. These compounds induce cell apoptosis, target PI3K/Akt/mTOR signaling pathways, inhibit carbonic anhydrase, and disrupt microtubules. Additionally, they inhibit tumor multidrug resistance and angiogenesis and regulate reactive oxygen species. Specific coumarin derivatives, such as auraptene, praeruptorin, osthole, and scopoletin, show anti-invasive, anti-migratory, and antiproliferative activities by arresting the cell cycle and inducing apoptosis. They also inhibit metalloproteinases-2 and -9, reducing tumor cell migration, invasion, and metastasis. These compounds can sensitize tumor cells to radiotherapy and chemotherapy. Synthetic coumarin derivatives also demonstrate potent antitumor and anticancer activities with minimal side effects. Given their diverse mechanisms of action and minimal side effects, coumarin-class phytochemicals hold significant potential as therapeutic agents in gynecological cancers, potentially improving treatment outcomes and reducing side effects. This review will aid in the synthesis and development of novel coumarin-based drugs for these cancers.
Collapse
Affiliation(s)
| | - Berrak Dumlupınar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Okan University, İstanbul, Türkiye
| | - Engin Celep
- Department of Pharmacognosy, Faculty of Pharmacy, Acıbadem Mehmet Ali Aydinlar University, Atasehir, Istanbul, Türkiye
| | - Inci Kurt Celep
- Department of Biotechnology, Faculty of Pharmacy, Istanbul Okan University, Istanbul, Türkiye
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado Facultad de Ciencias de la Salud Universidad Central de Chile, Santiago, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
7
|
Kaveh Zenjanab M, Hashemzadeh N, Alimohammadvand S, Sharifi-Azad M, Dalir Abdolahinia E, Jahanban-Esfahlan R. Notch Signaling Suppression by Golden Phytochemicals: Potential for Cancer Therapy. Adv Pharm Bull 2024; 14:302-313. [PMID: 39206407 PMCID: PMC11347744 DOI: 10.34172/apb.2024.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/09/2024] [Accepted: 03/03/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is one of the main causes of mortality worldwide. Cancer cells are characterized by unregulated cellular processes, including proliferation, progression, and angiogenesis. The occurrence of these processes is due to the dysregulation of various signaling pathways such as NF-κB (nuclear factor-κB), Wnt/beta-catenin, Notch signaling and MAPK (mitogen-activated protein kinases). Notch signaling pathways cause the progression of various types of malignant tumors. Among the phytochemicals for cancer therapy, several have attracted great interest, including curcumin, genistein, quercetin, silibinin, resveratrol, cucurbitacin and glycyrrhizin. Given the great cellular and molecular heterogeneity within tumors and the high toxicity and side effects of synthetic chemotherapeutics, natural products with pleiotropic effects that simultaneously target numerous signaling pathways appear to be ideal substitutes for cancer therapy. With this in mind, we take a look at the current status, impact and potential of known compounds as golden phytochemicals on key signaling pathways in tumors, focusing on the Notch pathway. This review may be useful for discovering new molecular targets for safe and efficient cancer therapy with natural chemotherapeutics.
Collapse
Affiliation(s)
| | - Nastaran Hashemzadeh
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Sharifi-Azad
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, US
| | - Rana Jahanban-Esfahlan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Li H, Sun J, Hu H, Wang Y. Transcription Factor E2F8 Activates PDK1-Mediated DNA Damage Repair to Enhance Cisplatin Resistance in Lung Adenocarcinoma. Pharmacology 2024; 109:341-356. [PMID: 38810606 DOI: 10.1159/000537819] [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: 08/10/2023] [Accepted: 02/12/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Cisplatin (DDP) is the commonest chemo drug in lung adenocarcinoma (LUAD) treatment, and DDP resistance is a significant barrier to therapeutic therapy. This study attempted to elucidate the impact of PDK1 on DDP resistance in LUAD and its mechanism. METHODS Bioinformatics analysis was used to determine the expression and enriched pathways of PDK1 in LUAD tissue. Subsequently, E2F8, the upstream transcription factor of PDK1, was predicted, and the binding relationship between the two was analyzed using dual-luciferase and ChIP experiments. PDK1 and E2F8 levels in LUAD tissues and cells were detected via qRT-PCR. Cell viability, proliferation, and apoptosis levels were assayed by CCK-8, EdU, and flow cytometry experiments, respectively. Comet assay was used to assess DNA damage, and immunofluorescence was used to assess the expression of γ-H2AX. NHEJ reporter assay was to assess DNA repair efficiency. Western blot tested levels of DNA damage repair (DDR)-related proteins. Immunohistochemistry assessed the expression of relevant genes. Finally, an animal model was constructed to investigate the influence of PDK1 expression on LUAD growth. RESULTS PDK1 was found to be upregulated in LUAD and enhanced DDP resistance by mediating DDR. E2F8 was identified as an upstream transcription factor of PDK1 and was highly expressed in LUAD. Rescue experiments presented that knocking down E2F8 could weaken the promotion of PDK1 overexpression on DDR-mediated DDP resistance in LUAD. In vivo experiments showed that knocking down PDK1 plus DDP significantly reduced the growth of xenograft tumors. CONCLUSION Our results indicated that the E2F8/PDK1 axis mediated DDR to promote DDP resistance in LUAD. Our findings lead to an improved treatment strategy after drug resistance.
Collapse
Affiliation(s)
- Hongliang Li
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Junxia Sun
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Haibo Hu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Yi Wang
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| |
Collapse
|
9
|
Kulavi S, Dhar D, Kamal IM, Chakrabarti S, Bandyopadhyay J. EIF4A3 targeted therapeutic intervention in glioblastoma multiforme using phytochemicals from Indian medicinal plants - an integration of phytotherapy into precision onco-medicine. J Biomol Struct Dyn 2024:1-21. [PMID: 38345073 DOI: 10.1080/07391102.2024.2314257] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/28/2024] [Indexed: 03/11/2025]
Abstract
Glioblastoma Multiforme (GBM), an aggressive brain tumor (grade-IV astrocytoma), poses treatment challenges. Poor prognosis results from the rapid growth, highlighting the role of EIF4A3 in regulating non-coding RNAs. EIF4A3 promotes the expression of several non-coding RNAs, viz, Circ matrix metallopeptidase 9 (MMP9), a prominent oncogene, by interacting with the upstream region of the circMMP9 mRNA transcript and acts on cell proliferation, migration, and invasion of GBM. However, research shows that EIF4A3 knockdown inhibits glioblastoma progression and increases apoptosis. In this study, we explored the efficiency of the phytochemicals from plants like Withania somnifera and Castanea sativa with potential anti-glioblastoma effects as obtained from the Indian Medicinal Plants, Phytochemistry and Therapeutics (IMPPAT) database. Consequently, we have performed a virtual screening of the compounds against the protein EIF4A3. We further investigated the efficiency of the shortlisted compounds based on docking scores evaluated using GOLD, AutoDock4.2, LeDock, and binding free energy analyses using Molecular Mechanics Poisson-Boltzmann Surface Area (MMPBSA). Among the phytochemicals studied so far, several Withania-specific compounds from Withania somnifera and a single dietary compound, viz., Thiamine from Castanea sativa, have exhibited comparatively good blood-brain barrier permeability, significant binding affinity towards the EIF4A3, and good ADMET properties. Furthermore, we have verified the interaction stability of the lead molecules with EIF4A3 using MD simulations. Thus, the present study offers an opportunity to develop drug candidates targeting glioblastoma caused by EIF4A3 over-expression, integrating phytotherapy into precision oncology to create tailored and precise natural treatment strategies for cancer.
Collapse
Affiliation(s)
- Sohini Kulavi
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia, India
| | - Debajit Dhar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia, India
| | - Izaz Monir Kamal
- Structural Biology and Bioinformatics Division, CSIR Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad, India
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad, India
| | - Jaya Bandyopadhyay
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia, India
| |
Collapse
|
10
|
Grover P, Thakur K, Bhardwaj M, Mehta L, Raina SN, Rajpal VR. Phytotherapeutics in Cancer: From Potential Drug Candidates to Clinical Translation. Curr Top Med Chem 2024; 24:1050-1074. [PMID: 38279745 DOI: 10.2174/0115680266282518231231075311] [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: 09/15/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 01/28/2024]
Abstract
Annually, a significant number of individuals succumb to cancer, an anomalous cellular condition characterized by uncontrolled cellular proliferation and the emergence of highly perilous tumors. Identifying underlying molecular mechanism(s) driving disease progression has led to various inventive therapeutic approaches, many of which are presently under pre-clinical and/or clinical trials. Over the recent years, numerous alternative strategies for addressing cancer have also been proposed and put into practice. This article delineates the modern therapeutic drugs employed in cancer treatment and their associated toxicity. Due to inherent drug toxicity associated with most modern treatments, demand rises for alternative therapies and phytochemicals with minimal side effects and proven efficacy against cancer. Analogs of taxol, Vinca alkaloids like vincristine and vinblastine, and podophyllotoxin represent a few illustrative examples in this context. The phytochemicals often work by modifying the activity of molecular pathways that are thought to be involved in the onset and progression of cancer. The principal objective of this study is to provide an overview of our current understanding regarding the pharmacologic effects and molecular targets of the active compounds found in natural products for cancer treatment and collate information about the recent advancements in this realm. The authors' interest in advancing the field of phytochemical research stems from both the potential of these compounds for use as drugs as well as their scientific validity. Accordingly, the significance of herbal formulations is underscored, shedding light on anticancer phytochemicals that are sought after at both pre-clinical and clinical levels, with discussion on the opportunities and challenges in pre-clinical and clinical cancer studies.
Collapse
Affiliation(s)
- Parul Grover
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | | | - Monika Bhardwaj
- Natural Product and Medicinal Chemistry Division, Indian Institute of Integrative Medicine (CSIR-IIIM), Jammu, 180001, India
| | - Lovekesh Mehta
- Amity Institute of Pharmacy, Amity University, Noida, 201301, India
| | - Soom Nath Raina
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, Noida, 201301, India
| | - Vijay Rani Rajpal
- Department of Botany, Hansraj College, Delhi University, Delhi, 110007, India
| |
Collapse
|
11
|
Li C, Liu FY, Shen Y, Tian Y, Han FJ. Research progress on the mechanism of glycolysis in ovarian cancer. Front Immunol 2023; 14:1284853. [PMID: 38090580 PMCID: PMC10715264 DOI: 10.3389/fimmu.2023.1284853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Glycolysis is the preferred energy metabolism pathway in cancer cells even when the oxygen content is sufficient. Through glycolysis, cancer cells convert glucose into pyruvic acid and then lactate to rapidly produce energy and promote cancer progression. Changes in glycolysis activity play a crucial role in the biosynthesis and energy requirements of cancer cells needed to maintain growth and metastasis. This review focuses on ovarian cancer and the significance of key rate-limiting enzymes (hexokinase, phosphofructokinase, and pyruvate kinase, related signaling pathways (PI3K-AKT, Wnt, MAPK, AMPK), transcription regulators (HIF-1a), and non-coding RNA in the glycolytic pathway. Understanding the relationship between glycolysis and these different mechanisms may provide new opportunities for the future treatment of ovarian cancer.
Collapse
Affiliation(s)
- Chan Li
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Fang-Yuan Liu
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Ying Shen
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Yuan Tian
- Zhejiang University of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Feng-Juan Han
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| |
Collapse
|
12
|
Ma Y, Lai X, Wen Z, Zhou Z, Yang M, Chen Q, Wang X, Mei F, Yang L, Yin T, Sun S, Lu G, Qi J, Lin H, Han H, Yang Y. Design, synthesis and biological evaluation of novel modified dual-target shikonin derivatives for colorectal cancer treatment. Bioorg Chem 2023; 139:106703. [PMID: 37399615 DOI: 10.1016/j.bioorg.2023.106703] [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: 05/14/2023] [Revised: 06/15/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
Warburg effect provides energy and material essential for tumor proliferation, the reverse of Warburg effect provides insights into the development of a novel anti-cancer strategy. Pyruvate kinase 2 (PKM2) and pyruvate dehydrogenase kinase 1 (PDK1) are two key enzymes in tumor glucose metabolism pathway that not only contribute to the Warburg effect through accelerating aerobic glycolysis, but also serve as druggable target for colorectal cancer (CRC). Considering that targeting PKM2 or PDK1 alone does not seem to be sufficient to remodel abnormal glucose metabolism and achieve significant antitumor activity, a series of novel benzenesulfonyl shikonin derivatives were designed to regulate PKM2 and PDK1 simultaneously. By means of molecular docking and antiproliferative screen, we found that compound Z10 could act as the combination of PKM2 activator and PDK1 inhibitor, thereby significantly inhibited glycolysis that reshaping tumor metabolism. Moreover, Z10 could inhibit proliferation, migration and induce apoptosis in CRC cell HCT-8. Finally, the in vivo anti-tumor activity of Z10 was evaluated in a colorectal cancer cell xenograft model in nude mice and the results demonstrated that Z10 induced tumor cell apoptosis and inhibited tumor cell proliferation with lower toxicity than shikonin. Our findings indicated that it is feasible to alter tumor energy metabolism through multi-target synergies, and the dual-target benzenesulfonyl shikonin derivative Z10 could be a potential anti-CRC agent.
Collapse
Affiliation(s)
- Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaohui Lai
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; School of Biology and Geography Science, Yili Normal University, Yining 835000, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Ziling Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Feng Mei
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Liu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Shucun Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Guihua Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Hongwei Han
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| | - Yonghua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
13
|
Soler Palacios B, Villares R, Lucas P, Rodríguez-Frade JM, Cayuela A, Piccirillo JG, Lombardía M, Delgado Gestoso D, Fernández-García M, Risco C, Barbas C, Corrales F, Sorzano COS, Martínez-Martín N, Conesa JJ, Iborra FJ, Mellado M. Growth hormone remodels the 3D-structure of the mitochondria of inflammatory macrophages and promotes metabolic reprogramming. Front Immunol 2023; 14:1200259. [PMID: 37475858 PMCID: PMC10354525 DOI: 10.3389/fimmu.2023.1200259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/14/2023] [Indexed: 07/22/2023] Open
Abstract
INTRODUCTION Macrophages are a heterogeneous population of innate immune cells that support tissue homeostasis through their involvement in tissue development and repair, and pathogen defense. Emerging data reveal that metabolism may control macrophage polarization and function and, conversely, phenotypic polarization may drive metabolic reprogramming. METHODS Here we use biochemical analysis, correlative cryogenic fluorescence microscopy and cryo-focused ion-beam scanning electron microscopy. RESULTS We demonstrate that growth hormone (GH) reprograms inflammatory GM-CSF-primed monocyte-derived macrophages (GM-MØ) by functioning as a metabolic modulator. We found that exogenous treatment of GM-MØ with recombinant human GH reduced glycolysis and lactate production to levels similar to those found in anti-inflammatory M-MØ. Moreover, GH treatment of GM-MØ augmented mitochondrial volume and altered mitochondrial dynamics, including the remodeling of the inner membrane to increase the density of cristae. CONCLUSIONS Our data demonstrate that GH likely serves a modulatory role in the metabolism of inflammatory macrophages and suggest that metabolic reprogramming of macrophages should be considered as a new target to intervene in inflammatory diseases.
Collapse
Affiliation(s)
- Blanca Soler Palacios
- Department of Immunology and Oncology, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Ricardo Villares
- Department of Immunology and Oncology, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Pilar Lucas
- Department of Immunology and Oncology, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - José Miguel Rodríguez-Frade
- Department of Immunology and Oncology, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Ana Cayuela
- Biocomputing Unit, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Jonathan G. Piccirillo
- Department of Macromolecular Structures, National Center for Biotechnology/The Spanish National Research Council) (CSIC), Madrid, Spain
| | - Manuel Lombardía
- Functional Proteomics Laboratory, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - David Delgado Gestoso
- Department of Macromolecular Structures, National Center for Biotechnology/The Spanish National Research Council) (CSIC), Madrid, Spain
| | - Miguel Fernández-García
- Metabolomic and Bioanalysis Center (CEMBIO), Pharmacy Faculty, Universidad San Pablo-CEU, Centre for Universitary Studies (CEU) Universities, Boadilla del Monte, Spain
- Department of Basic Medical Sciences, Medicine Faculty, Universidad San Pablo-CEU, Centre for Universitary Studies (CEU) Universities, Boadilla del Monte, Spain
| | - Cristina Risco
- Department of Macromolecular Structures, National Center for Biotechnology/The Spanish National Research Council) (CSIC), Madrid, Spain
| | - Coral Barbas
- Metabolomic and Bioanalysis Center (CEMBIO), Pharmacy Faculty, Universidad San Pablo-CEU, Centre for Universitary Studies (CEU) Universities, Boadilla del Monte, Spain
| | - Fernando Corrales
- Functional Proteomics Laboratory, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Carlos Oscar S. Sorzano
- Biocomputing Unit, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| | - Nuria Martínez-Martín
- Tissue and Organ Homeostasis Program, Centro de Biologia Molecular Severo Ochoa, The Spanish National Research Council (CSIC)–Autonomus University of Madrid (UAM), Madrid, Spain
| | - José Javier Conesa
- Department of Macromolecular Structures, National Center for Biotechnology/The Spanish National Research Council) (CSIC), Madrid, Spain
| | - Francisco J. Iborra
- Príncipe Felípe Research Centre (Associated Unit to the Biomedicine Institute of Valencia), Biomedicine Institute of Valencia, Valencia, Spain
| | - Mario Mellado
- Department of Immunology and Oncology, National Center for Biotechnology/The Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
14
|
Nazam N, Jabir NR, Ahmad I, Alharthy SA, Khan MS, Ayub R, Tabrez S. Phenolic Acids-Mediated Regulation of Molecular Targets in Ovarian Cancer: Current Understanding and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:274. [PMID: 37259418 PMCID: PMC9962268 DOI: 10.3390/ph16020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a global health concern with a dynamic rise in occurrence and one of the leading causes of mortality worldwide. Among different types of cancer, ovarian cancer (OC) is the seventh most diagnosed malignant tumor, while among the gynecological malignancies, it ranks third after cervical and uterine cancer and sadly bears the highest mortality and worst prognosis. First-line treatments have included a variety of cytotoxic and synthetic chemotherapeutic medicines, but they have not been particularly effective in extending OC patients' lives and are associated with side effects, recurrence risk, and drug resistance. Hence, a shift from synthetic to phytochemical-based agents is gaining popularity, and researchers are looking into alternative, cost-effective, and safer chemotherapeutic strategies. Lately, studies on the effectiveness of phenolic acids in ovarian cancer have sparked the scientific community's interest because of their high bioavailability, safety profile, lesser side effects, and cost-effectiveness. Yet this is a road less explored and critically analyzed and lacks the credibility of the novel findings. Phenolic acids are a significant class of phytochemicals usually considered in the nonflavonoid category. The current review focused on the anticancer potential of phenolic acids with a special emphasis on chemoprevention and treatment of OC. We tried to summarize results from experimental, epidemiological, and clinical studies unraveling the benefits of various phenolic acids (hydroxybenzoic acid and hydroxycinnamic acid) in chemoprevention and as anticancer agents of clinical significance.
Collapse
Affiliation(s)
- Nazia Nazam
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida 201301, Uttar Pradesh, India
| | - Nasimudeen R. Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur 613403, Tamil Nadu, India
| | - Iftikhar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Saif A. Alharthy
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashid Ayub
- Technology and Innovation Unit, Department of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
15
|
Silva VLM, Silva-Reis R, Moreira-Pais A, Ferreira T, Oliveira PA, Ferreira R, Cardoso SM, Sharifi-Rad J, Butnariu M, Costea MA, Grozea I. Dicoumarol: from chemistry to antitumor benefits. Chin Med 2022; 17:145. [PMID: 36575479 PMCID: PMC9793554 DOI: 10.1186/s13020-022-00699-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Dicoumarol, a coumarin-like compound, is known for its anticoagulant properties associated with the ability to inhibit vitamin K, being prescribed as a drug for several decades. The pharmaceutical value of dicoumarol turned it into a focus of chemists' attention, aiming its synthesis and of dicoumarol derivatives, bringing to light new methodologies. In recent years, several other bioactive effects have been claimed for dicoumarol and its derivatives, including anti-inflammatory, antimicrobial, antifungal, and anticancer, although the mechanisms of action underlying them are mostly not disclosed and additional research is needed to unravel them. This review presents a state of the art on the chemistry of dicoumarols, and their potential anticancer characteristics, highlighting the mechanisms of action elucidated so far. In parallel, we draw attention to the lack of in vivo studies and clinical trials to assess the safety and efficacy as drugs for later application.
Collapse
Affiliation(s)
- Vera L. M. Silva
- grid.7311.40000000123236065LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Silva-Reis
- grid.7311.40000000123236065LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Alexandra Moreira-Pais
- grid.7311.40000000123236065LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal ,grid.12341.350000000121821287Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal ,grid.5808.50000 0001 1503 7226Laboratory for Integrative and Translational Research in Population Health (ITR), Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP), 4200-450 Porto, Portugal
| | - Tiago Ferreira
- grid.12341.350000000121821287Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal ,grid.12341.350000000121821287Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Paula A. Oliveira
- grid.12341.350000000121821287Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal ,grid.12341.350000000121821287Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal ,grid.12341.350000000121821287Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Rita Ferreira
- grid.7311.40000000123236065LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Susana M. Cardoso
- grid.7311.40000000123236065LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Javad Sharifi-Rad
- grid.442126.70000 0001 1945 2902Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Monica Butnariu
- Life Sciences University “King Mihai I” from Timisoara, 300645 Calea Aradului 119, Timis, Romania
| | - Maria Alina Costea
- Life Sciences University “King Mihai I” from Timisoara, 300645 Calea Aradului 119, Timis, Romania
| | - Ioana Grozea
- Life Sciences University “King Mihai I” from Timisoara, 300645 Calea Aradului 119, Timis, Romania
| |
Collapse
|
16
|
Kobayashi H. Recent advances in understanding the metabolic plasticity of ovarian cancer: A systematic review. Heliyon 2022; 8:e11487. [PMID: 36406733 PMCID: PMC9668530 DOI: 10.1016/j.heliyon.2022.e11487] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecologic malignancy with a poor prognosis due to resistance to first-line chemotherapeutic agents. Some cancer cells are primarily dependent on glycolysis, but others favor mitochondrial oxidative phosphorylation (OXPHOS) over glycolysis. Changes in metabolic reprogramming have been reported to be involved in cancer cell survival. In this review, we summarize the metabolic profiles (e.g., metabolic heterogeneity, plasticity, and reprogramming) and adaptation to the dynamic tumor microenvironment and discuss potential novel therapeutic strategies. A literature search was performed between January 2000 and March 2022 in the PubMed and Google Scholar databases using a combination of specific terms. Ovarian cancer cells, including cancer stem cells, depend on glycolysis, OXPHOS, or both for survival. Several environmental stresses, such as nutrient starvation or glucose deprivation, hypoxic stress, acidification, and excessive reactive oxygen species (ROS) generation, reprogram the metabolic pathways to adapt. The interaction between tumors and adjacent stromal cells allows cancer cells to enhance mitochondrial energy metabolism. The metabolic reprogramming varies depending on genomic and epigenetic alterations of metabolism-related genes and the metabolic environment. Developing accurate and non-invasive methods for early identification of metabolic alterations could facilitate optimal cancer diagnosis and treatment. Cancer metabolism research has entered an exciting era where novel strategies targeting metabolic profiling will become more innovative.
Collapse
|
17
|
Lin Y, Zhou X, Ni Y, Zhao X, Liang X. Metabolic reprogramming of the tumor immune microenvironment in ovarian cancer: A novel orientation for immunotherapy. Front Immunol 2022; 13:1030831. [PMID: 36311734 PMCID: PMC9613923 DOI: 10.3389/fimmu.2022.1030831] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic tumor, with the highest mortality rate. Numerous studies have been conducted on the treatment of ovarian cancer in the hopes of improving therapeutic outcomes. Immune cells have been revealed to play a dual function in the development of ovarian cancer, acting as both tumor promoters and tumor suppressors. Increasingly, the tumor immune microenvironment (TIME) has been proposed and confirmed to play a unique role in tumor development and treatment by altering immunosuppressive and cytotoxic responses in the vicinity of tumor cells through metabolic reprogramming. Furthermore, studies of immunometabolism have provided new insights into the understanding of the TIME. Targeting or activating metabolic processes of the TIME has the potential to be an antitumor therapy modality. In this review, we summarize the composition of the TIME of ovarian cancer and its metabolic reprogramming, its relationship with drug resistance in ovarian cancer, and recent research advances in immunotherapy.
Collapse
|
18
|
Involvement of Phytochemical-Encapsulated Nanoparticles' Interaction with Cellular Signalling in the Amelioration of Benign and Malignant Brain Tumours. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113561. [PMID: 35684498 PMCID: PMC9182026 DOI: 10.3390/molecules27113561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/05/2022]
Abstract
Brain tumours have unresolved challenges that include delay prognosis and lower patient survival rate. The increased understanding of the molecular pathways underlying cancer progression has aided in developing various anticancer medications. Brain cancer is the most malignant and invasive type of cancer, with several subtypes. According to the WHO, they are classified as ependymal tumours, chordomas, gangliocytomas, medulloblastomas, oligodendroglial tumours, diffuse astrocytomas, and other astrocytic tumours on the basis of their heterogeneity and molecular mechanisms. The present study is based on the most recent research trends, emphasising glioblastoma cells classified as astrocytoma. Brain cancer treatment is hindered by the failure of drugs to cross the blood–brain barrier (BBB), which is highly impregnableto foreign molecule entry. Moreover, currently available medications frequently fail to cross the BBB, whereas chemotherapy and radiotherapy are too expensive to be afforded by an average incomeperson and have many associated side effects. When compared to our current understanding of molecularly targeted chemotherapeutic agents, it appears that investigating the efficacy of specific phytochemicals in cancer treatment may be beneficial. Plants and their derivatives are game changers because they are efficacious, affordable, environmentally friendly, faster, and less toxic for the treatment of benign and malignant tumours. Over the past few years, nanotechnology has made a steady progress in diagnosing and treating cancers, particularly brain tumours. This article discusses the effects of phytochemicals encapsulated in nanoparticles on molecular targets in brain tumours, along with their limitations and potential challenges.
Collapse
|
19
|
Alam M, Alam S, Shamsi A, Adnan M, Elasbali AM, Al-Soud WA, Alreshidi M, Hawsawi YM, Tippana A, Pasupuleti VR, Hassan MI. Bax/Bcl-2 Cascade Is Regulated by the EGFR Pathway: Therapeutic Targeting of Non-Small Cell Lung Cancer. Front Oncol 2022; 12:869672. [PMID: 35402265 PMCID: PMC8990771 DOI: 10.3389/fonc.2022.869672] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung carcinoma (NSCLC) comprises 80%-85% of lung cancer cases. EGFR is involved in several cancer developments, including NSCLC. The EGFR pathway regulates the Bax/Bcl-2 cascade in NSCLC. Increasing understanding of the molecular mechanisms of fundamental tumor progression has guided the development of numerous antitumor drugs. The development and improvement of rationally planned inhibitors and agents targeting particular cellular and biological pathways in cancer have been signified as a most important paradigm shift in the strategy to treat and manage lung cancer. Newer approaches and novel chemotherapeutic agents are required to accompany present cancer therapies for improving efficiency. Using natural products as a drug with an effective delivery system may benefit therapeutics. Naturally originated compounds such as phytochemicals provide crucial sources for novel agents/drugs and resources for tumor therapy. Applying the small-molecule inhibitors (SMIs)/phytochemicals has led to potent preclinical discoveries in various human tumor preclinical models, including lung cancer. In this review, we summarize recent information on the molecular mechanisms of the Bax/Bcl-2 cascade and EGFR pathway in NSCLC and target them for therapeutic implications. We further described the therapeutic potential of Bax/Bcl-2/EGFR SMIs, mainly those with more potent and selectivity, including gefitinib, EGCG, ABT-737, thymoquinone, quercetin, and venetoclax. In addition, we explained the targeting EGFR pathway and ongoing in vitro and in vivo and clinical investigations in NSCLC. Exploration of such inhibitors facilitates the future treatment and management of NSCLC.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, India
| | - Shoaib Alam
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, India
| | - Anas Shamsi
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Sakaka, Saudi Arabia
| | - Waleed Abu Al-Soud
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
- Health Sciences Research Unit, Jouf University, Sakaka, Saudi Arabia
| | - Mousa Alreshidi
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Hail, Hail, Saudi Arabia
| | | | - Anitha Tippana
- Regional Agricultural Research Station, Acharya N. G. Ranga Agricultural University (ANGRAU), Tirupati, India
| | - Visweswara Rao Pasupuleti
- Department of Biomedical Sciences and Therapeutics, Faculty of Medicine & Health Sciences, University Malaysia Sabah, Kota Kinabalu, Malaysia
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Pekanbaru, Indonesia
- Centre for International Collaboration and Research, Reva University, Rukmini Knowledge Park, Bangalore, India
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, India
| |
Collapse
|
20
|
Li W, Ding Z, Wang D, Li C, Pan Y, Zhao Y, Zhao H, Lu T, Xu R, Zhang S, Yuan B, Zhao Y, Yin Y, Gao Y, Li J, Yan M. Ten-gene signature reveals the significance of clinical prognosis and immuno-correlation of osteosarcoma and study on novel skeleton inhibitors regarding MMP9. Cancer Cell Int 2021; 21:377. [PMID: 34261456 PMCID: PMC8281696 DOI: 10.1186/s12935-021-02041-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES This study aimed to identify novel targets in the carcinogenesis, therapy and prognosis of osteosarcoma from genomic level, together with screening ideal lead compounds with potential inhibition regarding MMP-9. METHODS Gene expression profiles from GSE12865, GSE14359, GSE33382, GSE36001 and GSE99671 were obtained respectively from GEO database. Differentially expressed genes were identified, and functional enrichment analysis, such as GO, KEGG, GSEA, PPI were performed to make a comprehensive understanding of the hub genes. Next, a series of high-precision computational techniques were conducted to screen potential lead compounds targeting MMP9, including virtual screening, ADME, toxicity prediction, and accurate docking analysis. RESULTS 10 genes, MMP9, CD74, SPP1, CXCL12, TYROBP, FCER1G, HCLS1, ARHGDIB, LAPTM5 and IGF1R were identified as hub genes in the initiation of osteosarcoma. Machine learning, multivariate Cox analysis, ssGSEA and survival analysis demonstrated that these genes had values in prognosis, immune-correlation and targeted treatment. Tow novel compounds, ZINC000072131515 and ZINC000004228235, were screened as potential inhibitor regarding MMP9, and they could bind to MMP9 with favorable interaction energy and high binding affinity. Meanwhile, they were precited to be efficient and safe drugs with low-ames mutagenicity, none weight evidence of carcinogenicity, as well as non-toxic with liver. CONCLUSIONS This study revealed the significance of 10-gene signature in the development of osteosarcoma. Besides, drug candidates identified in this study provided a solid basis on MMP9 inhibitors' development.
Collapse
Affiliation(s)
- Weihang Li
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Ziyi Ding
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Dong Wang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chengfei Li
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Yikai Pan
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Yingjing Zhao
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Hongzhe Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Tianxing Lu
- Hou Zonglian Medical Experimental Class, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rui Xu
- Department of Endocrinology, Shanghai National Research Center for Endocrine and Metabolic Disease, State Key Laboratory of Medical Genomics, Shanghai Institute for Endocrine and Metabolic Disease, Ruijin Hospital. Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Shilei Zhang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Bin Yuan
- Department of Spine Surgery, Daxing Hospital, Xi'an, Shaanxi, China
| | - Yunlong Zhao
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Yanjiang Yin
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Gao
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China.
| | - Jing Li
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Ming Yan
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
21
|
Yue L, Ren Y, Yue Q, Ding Z, Wang K, Zheng T, Chen G, Chen X, Li M, Fan L. α-Lipoic Acid Targeting PDK1/NRF2 Axis Contributes to the Apoptosis Effect of Lung Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6633419. [PMID: 34211631 PMCID: PMC8211503 DOI: 10.1155/2021/6633419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/12/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
As an antioxidant, α-lipoic acid (LA) has attracted much attention to cancer research. However, the exact mechanism of LA in cancer progression control and prevention remains to be unclear. In this study, we demonstrated that α-lipoic acid has inhibitory effects on the proliferation, migration, and proapoptotic effects of non-small-cell lung cancer (NSCLC) cell lines A549 and PC9. LA-induced NSCLC cell apoptosis was mediated by elevated mitochondrial reactive oxygen species (ROS). Further study confirmed that it is by downregulating the expression of PDK1 (the PDH kinase), resulted in less phospho-PDH phenotype which could interact with Keap1, the negative controller of NRF2, directly leading to NRF2 decrease. Thus, by downregulating the NRF2 antioxidant system, LA plays a role in promoting apoptosis through the ROS signaling pathway. Moreover, LA could enhance other PDK inhibitors with the proapoptosis effect. In summary, our study shows that LA promotes apoptosis and exerts its antitumor activity against lung cancer by regulating mitochondrial energy metabolism enzyme-related antioxidative stress system. Administration of LA to the tumor-bearing animal model further supported the antitumor effect of LA. These findings provided new ideas for the clinical application of LA in the field of cancer therapy.
Collapse
Affiliation(s)
- Liduo Yue
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Yanbei Ren
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Qingxi Yue
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 201999 Shanghai, China
| | - Zhou Ding
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Kai Wang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Guojie Chen
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Xiangyun Chen
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Ming Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Lihong Fan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| |
Collapse
|
22
|
Anwar S, Shamsi A, Mohammad T, Islam A, Hassan MI. Targeting pyruvate dehydrogenase kinase signaling in the development of effective cancer therapy. Biochim Biophys Acta Rev Cancer 2021; 1876:188568. [PMID: 34023419 DOI: 10.1016/j.bbcan.2021.188568] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Pyruvate is irreversibly decarboxylated to acetyl coenzyme A by mitochondrial pyruvate dehydrogenase complex (PDC). Decarboxylation of pyruvate is considered a crucial step in cell metabolism and energetics. The cancer cells prefer aerobic glycolysis rather than mitochondrial oxidation of pyruvate. This attribute of cancer cells allows them to sustain under indefinite proliferation and growth. Pyruvate dehydrogenase kinases (PDKs) play critical roles in many diseases because they regulate PDC activity. Recent findings suggest an altered metabolism of cancer cells is associated with impaired mitochondrial function due to PDC inhibition. PDKs inhibit the PDC activity via phosphorylation of the E1a subunit and subsequently cause a glycolytic shift. Thus, inhibition of PDK is an attractive strategy in anticancer therapy. This review highlights that PDC/PDK axis could be implicated in cancer's therapeutic management by developing potential small-molecule PDK inhibitors. In recent years, a dramatic increase in the targeting of the PDC/PDK axis for cancer treatment gained an attention from the scientific community. We further discuss breakthrough findings in the PDC-PDK axis. In addition, structural features, functional significance, mechanism of activation, involvement in various human pathologies, and expression of different forms of PDKs (PDK1-4) in different types of cancers are discussed in detail. We further emphasized the gene expression profiling of PDKs in cancer patients to prognosis and therapeutic manifestations. Additionally, inhibition of the PDK/PDC axis by small molecule inhibitors and natural compounds at different clinical evaluation stages has also been discussed comprehensively.
Collapse
Affiliation(s)
- Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Anas Shamsi
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
23
|
Pyruvate dehydrogenase kinases (PDKs): an overview toward clinical applications. Biosci Rep 2021; 41:228121. [PMID: 33739396 PMCID: PMC8026821 DOI: 10.1042/bsr20204402] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Pyruvate dehydrogenase kinase (PDK) can regulate the catalytic activity of pyruvate decarboxylation oxidation via the mitochondrial pyruvate dehydrogenase complex, and it further links glycolysis with the tricarboxylic acid cycle and ATP generation. This review seeks to elucidate the regulation of PDK activity in different species, mainly mammals, and the role of PDK inhibitors in preventing increased blood glucose, reducing injury caused by myocardial ischemia, and inducing apoptosis of tumor cells. Regulations of PDKs expression or activity represent a very promising approach for treatment of metabolic diseases including diabetes, heart failure, and cancer. The future research and development could be more focused on the biochemical understanding of the diseases, which would help understand the cellular energy metabolism and its regulation by pharmacological effectors of PDKs.
Collapse
|
24
|
Xu H, Li D, Ma J, Zhao Y, Xu L, Tian R, Liu Y, Sun L, Su J. The IL-33/ST2 axis affects tumor growth by regulating mitophagy in macrophages and reprogramming their polarization. Cancer Biol Med 2021; 18:172-183. [PMID: 33628592 PMCID: PMC7877183 DOI: 10.20892/j.issn.2095-3941.2020.0211] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/14/2020] [Indexed: 01/03/2023] Open
Abstract
Objective: Macrophages are a major component of the tumor microenvironment. M1 macrophages secrete pro-inflammatory factors that inhibit tumor growth and development, whereas tumor-associated macrophages (TAMs) mainly exhibit an M2 phenotype. Our previous studies have shown that the interleukin-33/ST2 (IL-33/ST2) axis is essential for activation of the M1 phenotype. This study investigates the role of the IL-33/ST2 axis in TAMs, its effects on tumor growth, and whether it participates in the mutual conversion between the M1 and M2 phenotypes. Methods: Bone marrow-derived macrophages were extracted from wildtype, ST2 knockout (ST2−/−), and Il33-overexpressing mice and differentiated with IL-4. The mitochondrial and lysosomal number and location, and the expression of related proteins were used to analyze mitophagy. Oxygen consumption rates and glucose and lactate levels were measured to reveal metabolic changes. Results: The IL-33/ST2 axis was demonstrated to play an important role in the metabolic conversion of macrophages from OXPHOS to glycolysis by altering mitophagy levels. The IL-33/ST2 axis promoted enhanced cell oxidative phosphorylation, thereby further increasing M2 polarization gene expression and ultimately promoting tumor growth (P < 0.05) (Figure 4). This metabolic shift was not due to mitochondrial damage, because the mitochondrial membrane potential was not significantly altered by IL-4 stimulation or ST2 knockout; however, it might be associated with the mTOR activity. Conclusions: These results clarify the interaction between the IL-33/ST2 pathway and macrophage polarization, and may pave the way to the development of new cancer immunotherapies targeting the IL-33/ST2 axis.
Collapse
Affiliation(s)
- Huadan Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Dong Li
- Department of Hepatology, The First Hospital of Jilin University, Changchun 130000, China.,Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Jiaoyan Ma
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Long Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Rui Tian
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| |
Collapse
|
25
|
Yao S, Shang W, Huang L, Xu R, Wu M, Wang F. The oncogenic and prognostic role of PDK1 in the progression and metastasis of ovarian cancer. J Cancer 2021; 12:630-643. [PMID: 33403023 PMCID: PMC7778543 DOI: 10.7150/jca.47278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal of gynecological tumors in women. Tumor metabolism has become a new opportunity in the treatment of tumors. Pyruvate dehydrogenase kinase 1 (PDK1), as a key regulatory enzyme implicated in metabolic reprogramming of tumors, abnormally high expressed in various tumors and involved in the regulation of tumor cell biological behavior. However, the role of PDK1 in the occurrence and development of ovarian cancer remains unclear. Our team identified the expression of PDK1 in ovarian cancer cell lines and tissues through RT-PCR and immunohistochemical staining and evaluated the correlation of PDK1 expression with clinicopathologic features of patients and survival analyses. We used a variety of in vitro experiments to explore the influence of PDK1 on proliferation, invasion, migration, colony formation, apoptosis and the cell cycle of ovarian cancer cell lines CAOV3 and SKOV3. PDK1 was highly expressed in ovarian cancer cell lines and OC tissues. High expression of PDK1 was closely correlated to tumor size, FIGO stage, extraovarian metastases status and distribution. Univariate and multivariate Cox regression analysis identified that PDK1 was an independent prognostic factor for overall survival. Moreover, PDK1 was a superior predictor in prognosis of ovarian cancer and the incorporation of CA125 into PDK1 generated a predictive combination that displayed better predictive accuracy for overall survival. Downregulation of PDK1 suppressed the biological behavior of ovarian cancer cells due to S phase arrest and cellular apoptosis. PDK1 may serve as a novel prognostic biomarker, even a promising antineoplastic target of ovarian cancer.
Collapse
Affiliation(s)
- Shasha Yao
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China.,Department of Laboratory Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, 211100, Nanjing, China
| | - Wenwen Shang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Lei Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Rui Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Ming Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| |
Collapse
|
26
|
Saidijam M, Afshar S, Taherkhani A. Identifying Potential Biomarkers in Colorectal Cancer and Developing Non-invasive Diagnostic Models Using Bioinformatics Approaches. AVICENNA JOURNAL OF MEDICAL BIOCHEMISTRY 2020. [DOI: 10.34172/ajmb.2020.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most frequent causes of gastrointestinal tumors. Due to the invasiveness of the current diagnostic methods, there is an urgent need to develop non-invasive diagnostic approaches for CRC. The exact mechanisms and the most important genes associated with the development of CRC are not fully demonstrated. Objectives: This study aimed to identify differentially expressed miRNAs (DEMs), key genes, and their regulators associated with the pathogenesis of CRC. The signaling pathways and biological processes (BPs) that were significantly affected in CRC were also indicated. Moreover, two non-invasive models were constructed for CRC diagnosis. Methods: The miRNA dataset GSE59856 was downloaded from the Gene Expression Omnibus (GEO) database and analyzed to identify DEMs in CRC patients compared with healthy controls (HCs). A protein-protein interaction (PPI) network was built and analyzed. Significant clusters in the PPI networks were identified, and the BPs and pathways associated with these clusters were studied. The hub genes in the PPI network, as well as their regulators were identified. Results: A total of 569 DEMs were demonstrated with the criteria of P value <0.001. A total of 110 essential genes and 30 modules were identified in the PPI network. Functional analysis revealed that 1005 BPs, 9 molecular functions (MFs), 14 cellular components (CCs), and 887 pathways were significantly affected in CRC. A total of 22 transcription factors (TFs) were demonstrated as the regulators of the hubs. Conclusion: Our results may provide new insight into the pathogenesis of CRC and advance the diagnostic and therapeutic methods of the disease. However, confirmation is required in the future.
Collapse
Affiliation(s)
- Massoud Saidijam
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
27
|
Chota A, George BP, Abrahamse H. Potential Treatment of Breast and Lung Cancer Using Dicoma anomala, an African Medicinal Plant. Molecules 2020; 25:molecules25194435. [PMID: 32992537 PMCID: PMC7582250 DOI: 10.3390/molecules25194435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022] Open
Abstract
Globally, cancer has been identified as one of the leading causes of death in public health. Its etiology is based on consistent exposure to carcinogenic. Plant-derived anticancer compounds are known to be less toxic to the normal cells and are classified into acetylenic compounds, phenolics, terpenes, and phytosterols. Dicoma anomala is a perennial herb belonging to the family Asteraceae and is widely distributed in Sub-Saharan Africa and used in the treatment of cancer, malaria, fever, diabetes, ulcers, cold, and cough. This review aimed at highlighting the benefits of D. anomala in various therapeutic applications with special reference to the treatment of cancers and the mechanisms through which the plant-derived agents induce cell death.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Correspondence: ; Tel.: +27-11-559-6550; Fax: +27-11-559-6448
| |
Collapse
|
28
|
McMullen M, Madariaga A, Lheureux S. New approaches for targeting platinum-resistant ovarian cancer. Semin Cancer Biol 2020; 77:167-181. [DOI: 10.1016/j.semcancer.2020.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
|
29
|
Xu H, He Y, Ma J, Zhao Y, Liu Y, Sun L, Su J. Inhibition of pyruvate dehydrogenase kinase‑1 by dicoumarol enhances the sensitivity of hepatocellular carcinoma cells to oxaliplatin via metabolic reprogramming. Int J Oncol 2020; 57:733-742. [PMID: 32705170 PMCID: PMC7384842 DOI: 10.3892/ijo.2020.5098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/22/2020] [Indexed: 01/02/2023] Open
Abstract
The Warburg effect is a unique metabolic feature of the majority of tumor cells and is closely related to chemotherapeutic resistance. Pyruvate dehydrogenase kinase 1 (PDK1) is considered a 'switch' that controls the fate of pyruvate in glucose metabolism. However, to date, to the best of our knowledge, there are only a few studies to available which had studied the reduction of chemotherapeutic resistance via the metabolic reprogramming of tumor cells with PDK1 as a target. In the present study, it was found dicoumarol (DIC) reduced the phosphorylation of pyruvate dehydrogenase (PDH) by inhibiting the activity of PDK1, which converted the metabolism of human hepatocellular carcinoma (HCC) cells to oxidative phosphorylation, leading to an increase in mitochondrial reactive oxygen species ROS (mtROS) and a decrease in mitochondrial membrane potential (MMP), thereby increasing the apoptosis induced by oxaliplatin (OXA). Furthermore, the present study elucidated that the targeting of PDK1 may be a potential strategy for targeting metabolism in the chemotherapy of HCC. In addition, DIC as an 'old drug' exhibits novel efficacy, bringing new hope for antitumor therapy.
Collapse
Affiliation(s)
- Huadan Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yichun He
- Department of Neurosurgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiaoyan Ma
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
30
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Kalantari M, Mohammadinejad R, Javaheri T, Sethi G. Association of the Epithelial-Mesenchymal Transition (EMT) with Cisplatin Resistance. Int J Mol Sci 2020; 21:E4002. [PMID: 32503307 PMCID: PMC7312011 DOI: 10.3390/ijms21114002] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Therapy resistance is a characteristic of cancer cells that significantly reduces the effectiveness of drugs. Despite the popularity of cisplatin (CP) as a chemotherapeutic agent, which is widely used in the treatment of various types of cancer, resistance of cancer cells to CP chemotherapy has been extensively observed. Among various reported mechanism(s), the epithelial-mesenchymal transition (EMT) process can significantly contribute to chemoresistance by converting the motionless epithelial cells into mobile mesenchymal cells and altering cell-cell adhesion as well as the cellular extracellular matrix, leading to invasion of tumor cells. By analyzing the impact of the different molecular pathways such as microRNAs, long non-coding RNAs, nuclear factor-κB (NF-ĸB), phosphoinositide 3-kinase-related protein kinase (PI3K)/Akt, mammalian target rapamycin (mTOR), and Wnt, which play an important role in resistance exhibited to CP therapy, we first give an introduction about the EMT mechanism and its role in drug resistance. We then focus specifically on the molecular pathways involved in drug resistance and the pharmacological strategies that can be used to mitigate this resistance. Overall, we highlight the various targeted signaling pathways that could be considered in future studies to pave the way for the inhibition of EMT-mediated resistance displayed by tumor cells in response to CP exposure.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran;
- Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran
| | - Mahshad Kalantari
- Department of Genetic Science, Tehran Medical Science Branch, Islamic Azad University, Tehran 19168931813, Iran;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 1355576169, Iran
| | - Tahereh Javaheri
- Health Informatics Lab, Metropolitan College, Boston University, Boston, MA 02215, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| |
Collapse
|
31
|
Han J, Yang D, Hall DR, Liu J, Sun J, Gu W, Tang S, Alharbi HA, Jones PD, Krause HM, Peng H. Toxicokinetics of Brominated Azo Dyes in the Early Life Stages of Zebrafish ( Danio rerio) Is Prone to Aromatic Substituent Changes. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:4421-4431. [PMID: 32146810 DOI: 10.1021/acs.est.9b07178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Brominated azo dyes (BADs) have been identified as predominant indoor brominated pollutants in daycare dust; thus, their potential health risk to children is of concern. However, the toxicities of BADs remain elusive. In this study, the toxicokinetics of two predominant BADs, Disperse Blue 373 (DB373) and Disperse Violet 93 (DV93), and their suspect metabolite 2-bromo-4,6-dinitroaniline (BDNA) was investigated in embryos of zebrafish (Danio rerio). The bioconcentration factor of DV93 at 120 hpf is 6.2-fold lower than that of DB373. The nontarget analysis revealed distinct metabolism routes between DB373 and DV93 by reducing nitro groups to nitroso (DB373) or amine (DV93), despite their similar structures. NAD(P)H quinone oxidoreductase 1 (NQO1) and pyruvate dehydrogenase were predicted as the enzymes responsible for the reduction of DB373 and DV93 by correlating time courses of the metabolites and enzyme development. Further in vitro recombinant enzyme and in vivo inhibition results validated NQO1 as the enzyme specifically reducing DB373, but not DV93. Global proteome profiling revealed that the expression levels of proteins from the "apoptosis-induced DNA fragmentation" pathway were significantly upregulated by all three BADs, supporting the bioactivation of BADs to mutagenic aromatic amines. This study discovered the bioactivation of BADs via distinct eukaryotic enzymes, implying their potential health risks.
Collapse
Affiliation(s)
- Jiajun Han
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Diwen Yang
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - David Ross Hall
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
- School of the Environment, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Jiabao Liu
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jianxian Sun
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Wen Gu
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Song Tang
- Department of Environmental Toxicology, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Hattan A Alharbi
- Department of Plant Protection, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia
| | - Paul D Jones
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
| | - Henry M Krause
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
- School of the Environment, University of Toronto, Toronto, ON M5S 3E8, Canada
| |
Collapse
|
32
|
PDK1 promotes ovarian cancer metastasis by modulating tumor-mesothelial adhesion, invasion, and angiogenesis via α5β1 integrin and JNK/IL-8 signaling. Oncogenesis 2020; 9:24. [PMID: 32071289 PMCID: PMC7028730 DOI: 10.1038/s41389-020-0209-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancies owing to the lack of definitive symptoms until development of widespread metastases. Identification of novel prognostic and therapeutic targets is therefore an urgent need to improve survival. Here, we demonstrated high expression of the mitochondrial gatekeeping enzyme, pyruvate dehydrogenase kinase 1 (PDK1), in both clinical samples and cell lines of ovarian cancer. PDK1 expression was significantly associated with metastasis, reduced chemosensitivity, and poor overall and disease-free survival, and further highlighted as an independent prognostic factor. Silencing of PDK1 retarded lactate production, ovarian cancer cell adhesion, migration, invasion, and angiogenesis, and consequently metastasis, concomitant with decreased α5β1 integrin expression. Phospho-kinase array profiling and RNA sequencing analyses further revealed reduction of JNK activation and IL-8 expression in PDK1-depleted cells. Conversely, PDK1 overexpression promoted cell adhesion via modulation of α5β1 integrins, along with cell migration, invasion, and angiogenesis through activation of JNK/IL-8 signaling. PDK1 depletion additionally hindered tumor growth and dissemination in nude mice in vivo. Importantly, PDK1 levels were upregulated upon treatment with conditioned medium from omental tissues, which in turn promoted metastasis. Our findings suggest that PDK1, which is regulated by the tumor microenvironment, controls lactate production and promotes ovarian cancer cell metastasis via modulation of α5β1 integrin and JNK/IL-8 signaling. To our knowledge, this is the first report to demonstrate an association between PDK1 and survival in patients with ovarian cancer, supporting its efficacy as a valuable prognostic marker and therapeutic molecular target for the disease.
Collapse
|
33
|
Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in Cancer Treatment: From Preclinical Studies to Clinical Practice. Front Pharmacol 2020; 10:1614. [PMID: 32116665 PMCID: PMC7025531 DOI: 10.3389/fphar.2019.01614] [Citation(s) in RCA: 518] [Impact Index Per Article: 103.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/10/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is a severe health problem that continues to be a leading cause of death worldwide. Increasing knowledge of the molecular mechanisms underlying cancer progression has led to the development of a vast number of anticancer drugs. However, the use of chemically synthesized drugs has not significantly improved the overall survival rate over the past few decades. As a result, new strategies and novel chemoprevention agents are needed to complement current cancer therapies to improve efficiency. Naturally occurring compounds from plants known as phytochemicals, serve as vital resources for novel drugs and are also sources for cancer therapy. Some typical examples include taxol analogs, vinca alkaloids such as vincristine, vinblastine, and podophyllotoxin analogs. These phytochemicals often act via regulating molecular pathways which are implicated in growth and progression of cancer. The specific mechanisms include increasing antioxidant status, carcinogen inactivation, inhibiting proliferation, induction of cell cycle arrest and apoptosis; and regulation of the immune system. The primary objective of this review is to describe what we know to date of the active compounds in the natural products, along with their pharmacologic action and molecular or specific targets. Recent trends and gaps in phytochemical based anticancer drug discovery are also explored. The authors wish to expand the phytochemical research area not only for their scientific soundness but also for their potential druggability. Hence, the emphasis is given to information about anticancer phytochemicals which are evaluated at preclinical and clinical level.
Collapse
Affiliation(s)
- Amit S Choudhari
- Combi-Chem Bio-Resource Center, Organic Chemistry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Pallavi C Mandave
- Interactive Research School of Health Affairs, Bharati Vidyapeeth Deemed University, Pune, India
| | - Manasi Deshpande
- Department of Dravyaguna Vigan, Ayurved Pharmacology, College of Ayurved, Bharati Vidyapeeth Deemed University, Pune, India
| | | | - Om Prakash
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
34
|
Kuznietsova H, Dziubenko N, Byelinska I, Hurmach V, Bychko A, Lynchak O, Milokhov D, Khilya O, Rybalchenko V. Pyrrole derivatives as potential anti-cancer therapeutics: synthesis, mechanisms of action, safety. J Drug Target 2019; 28:547-563. [PMID: 31814456 DOI: 10.1080/1061186x.2019.1703189] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Pyrrole derivatives (PDs) chloro-1-(4-chlorobenzyl)-4-((3-(trifluoromethyl)phenyl)amino)-1H-pyrrole-2,5-dione (MI-1) and 5-amino-4-(1,3-benzothyazol-2-yn)-1-(3-methoxyphenyl)-1,2-dihydro-3H-pyrrole-3-one (D1) were synthesised as inhibitors of several protein kinases including EGFR and VEGFR. The aim of the study was to reveal the exact mechanisms of PDs' action EGFR and VEGFR are involved in. We observed, that both PDs could bind with EGFR and VEGFR and form stable complexes. PDs entered into electrostatic interactions with polar groups of phospholipid heads in cell membrane, and the power of interaction depended on the nature of PD radical substituents (greater for MI-1 and smaller for D1). Partial intercalation of MI-1 into the membrane hydrophobic zone also occurred. PDs concentrations induced apoptosis in malignant cells but normal ones had different sensitivity to those. MI-1 and D1 acted like antioxidants in inflamed colonic tissue, as evidenced by reduce of lipid and protein peroxidation products (by 43-67%) and increase of superoxide dismutase activity (by 40 and 58%) with restoring these values to control ones. MI-1 restored reduced haemoglobin and normalised elevated platelets and monocytes in settings of colorectal cancer, whereas D1 normalised only platelets. Thus, MI-1 and D1 could be used as competitive inhibitors of EGFR and VEGFR and antioxidants, which might contribute to realisation of their anti-inflammatory, proapoptotic and antitumor activity.
Collapse
Affiliation(s)
| | | | | | - Vasyl Hurmach
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Andriy Bychko
- Bogomolets National Medical University, Kyiv, Ukraine
| | - Oksana Lynchak
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Demyd Milokhov
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olga Khilya
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | |
Collapse
|
35
|
Al-Alem LF, Baker AT, Pandya UM, Eisenhauer EL, Rueda BR. Understanding and Targeting Apoptotic Pathways in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11111631. [PMID: 31652965 PMCID: PMC6893837 DOI: 10.3390/cancers11111631] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer cells evade the immune system as well as chemotherapeutic and/or biologic treatments through inherent or acquired mechanisms of survival and drug resistance. Depending on the cell type and the stimuli, this threshold can range from external forces such as blunt trauma to programmed processes such as apoptosis, autophagy, or necroptosis. This review focuses on apoptosis, which is one form of programmed cell death. It highlights the multiple signaling pathways that promote or inhibit apoptosis and reviews current clinical therapies that target apoptotic pathways in ovarian cancer.
Collapse
Affiliation(s)
- Linah F Al-Alem
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Obstetrics and Gynecology, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew T Baker
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Obstetrics and Gynecology, Harvard Medical School, Boston, MA 02115, USA.
| | - Unnati M Pandya
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Obstetrics and Gynecology, Harvard Medical School, Boston, MA 02115, USA.
| | - Eric L Eisenhauer
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Obstetrics and Gynecology, Harvard Medical School, Boston, MA 02115, USA.
- Gynecology and Oncology Division, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Obstetrics and Gynecology, Harvard Medical School, Boston, MA 02115, USA.
- Gynecology and Oncology Division, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
36
|
Mayank, Singh A, Kaur N, Garg N, Singh N. Anticancer SAR establishment and novel accruing signal transduction model of drug action using biscoumarin scaffold. Comput Biol Chem 2019; 83:107104. [PMID: 31546212 DOI: 10.1016/j.compbiolchem.2019.107104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 01/20/2023]
Abstract
In this paper, we have established methylenebis (4-hydroxy-2H-chromen-2-one) as a promising anticancer scaffold with kinesin spindle protein (KSP) inhibitory activity under malignant condition. A series of biscoumarin derivatives (MN1 to MN30) with different substituent were synthesized, and their anticancer activity was explored. Six biscoumarin derivatives that were found active were further selected to formulate organic nanoparticles (ONPs). Anticancer activity of both the forms (viz conventional and ONPs) was compared. MN30 was found most potent whereby MN10 showed good anticancer activity in both, i.e., conventional and ONP form; the structural activity relationship (SAR) study has been established. Computational investigation revealed biscoumarin scaffold as a suitable pharmacophore to bind against KSP protein. Molecular dynamics simulation studies revealed protein-ligand stability and dynamic behavior of biscoumarin-KSP complex. Finally, accruing signal transduction model was formulated to explain the observed MTT trend of conventional and ONP form. The model seems useful towards solving population specific varied results of chemotherapeutic agents. According to the model, MN10 and MN30 derivatives have good pharmacodynamics inertia and therefore, both the molecules were able to provide dose-dependent cytotoxic results.
Collapse
Affiliation(s)
- Mayank
- Department of Chemistry, Indian Institute of Technology Ropar, Punjab, 140001, India
| | - Ashutosh Singh
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Navneet Kaur
- Department of Chemistry, Punjab University, Chandigarh, 160014, India.
| | - Neha Garg
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India.
| | - Narinder Singh
- Department of Chemistry, Indian Institute of Technology Ropar, Punjab, 140001, India.
| |
Collapse
|
37
|
The double inhibition of PDK1 and STAT3-Y705 prevents liver metastasis in colorectal cancer. Sci Rep 2019; 9:12973. [PMID: 31506552 PMCID: PMC6736869 DOI: 10.1038/s41598-019-49480-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
As a key glycolysis enzyme, the significance of pyruvate dehydrogenase kinase 1 (PDK1) in the development of colorectal cancer (CRC) remains unknown. This study revealed that the prognosis of CRC patients with high levels of PDK1 was poor, and PDK1 knockdown significantly reduced liver metastasis of CRC in both nude mice and immune competent BALB/C mice. When combined with cryptotanshinone (CPT), an inhibitor of STAT3-p-Y705, the liver metastasis was further inhibited. PDK1 knockdown obviously increased reactive oxygen species level in anoikis conditions and subsequently resulted in an elevated anoikis, but the combination of PDK1 knockdown and CPT showed a reduced effect on anoikis. Based on this discrepancy, the adherence ability of CRC cells to matrix protein fibronectin was further detected. It showed that PDK1 knockdown significantly decreased the adherence of CRC cells to fibronectin when combined with CPT. These results suggest that inhibition of PDK1 can decrease the surviving CRC cells in blood circulation via up-regulation of anoikis, and inhibition of STAT3-p-Y705 can prevent it to settle down on the liver premetastatic niche, which ultimately reduces liver metastasis.
Collapse
|
38
|
He J, Yu S, Guo C, Tan L, Song X, Wang M, Wu J, Long Y, Gong D, Zhang R, Cao Z, Li Y, Peng C. Polyphyllin I induces autophagy and cell cycle arrest via inhibiting PDK1/Akt/mTOR signal and downregulating cyclin B1 in human gastric carcinoma HGC-27 cells. Biomed Pharmacother 2019; 117:109189. [PMID: 31387191 DOI: 10.1016/j.biopha.2019.109189] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/17/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022] Open
Abstract
Paris polyphylla. is a traditional medicinal herb that has long been used to prevent cancer in many Asian countries. Polyphyllin I (PPI), an important bioactive constituent of Paris polyphylla, has been found to exhibit a wide variety of anticancer activities in many types of cancer cells. However, the effects of PPI on human gastric carcinoma cells and its mechanism of action remain unclear. In this study, we examined the effective anti-gastric carcinoma activity of PPI and its underlying mechanism of action in HGC-27 cells. In vitro, sub-micromolar concentrations of PPI inhibited HGC-27 cell proliferation with an IC50 of 0.34 ± 0.06 μM after a 72-h treatment. In vivo, 3 mg/kg PPI significantly inhibited proliferation of HGC-27 tumor cells, with a 78.8% inhibition rate compared to paclitaxel, and demonstrated higher safety. Analysis of MDC and mGFP-LC3 fluorescence, Western blotting and flow cytometry indicated that PPI induced cell cycle arrest in HGC-27 cells by promoting the conversion of LC3-I to LC3-II and by downregulating cyclin B1. Furthermore, Western blotting showed that PPI inhibited the autophagy-regulating PDK1/Akt/mTOR signaling pathway in vitro and in vivo. In addition, immunohistochemistry and TUNEL staining revealed that PPI decreased Ki67 expression and increased the percentage of apoptotic cells in HGC-27 xenograft tumors. These data indicate that PPI is an PDK1/Akt/mTOR signaling inhibitor and of therapeutic relevance for gastric cancer treatment and that the rhizome of Paris polyphylla deserves further clinical investigation as an alternative therapy for gastric cancer.
Collapse
Affiliation(s)
- Junlin He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Si Yu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China; Chengdu Medical College, Chengdu 610500, China
| | - Chuanjie Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Lu Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Xiaominting Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Miao Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Jing Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Yuling Long
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Daoyin Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Ruoqi Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Zhixing Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China
| | - Yuzhi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China.
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, China.
| |
Collapse
|
39
|
Xu H, Sun L, He Y, Yuan X, Niu J, Su J, Li D. Deficiency in IL-33/ST2 Axis Reshapes Mitochondrial Metabolism in Lipopolysaccharide-Stimulated Macrophages. Front Immunol 2019; 10:127. [PMID: 30774633 PMCID: PMC6367255 DOI: 10.3389/fimmu.2019.00127] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 01/15/2019] [Indexed: 12/26/2022] Open
Abstract
The polarization and function of macrophages play essential roles in controlling immune responses. Interleukin (IL)-33 is a member of the IL-1 family that has been shown to influence macrophage activation and polarization, but the underlying mechanisms are not fully understood. Mitochondrial metabolism has been reported to be a central player in shaping macrophage polarization; previous studies have shown that both aerobic glycolysis and oxidative phosphorylation uniquely regulate the functions of M1 and M2 macrophages. Whether IL-33 polarizes macrophages by reshaping mitochondrial metabolism requires further investigation. In this work, we examined the mitochondrial metabolism of bone marrow-derived macrophages (BMDMs) from either wild type (WT), Il33-overexpressing, or IL-33 receptor knockout (St2−/−) mice challenged with lipopolysaccharide (LPS). We found that after LPS stimulation, compared with WT BMDMs, St2−/− BMDMs had reduced cytokine production and increased numbers and activity of mitochondria via the metabolism regulator peroxisome proliferator-activated receptor-C coactivator-1 α (PGC1α). This was demonstrated by increased mitochondrial DNA copy number, mitochondria counts, mitochondria fission- and fusion-related gene expression, oxygen consumption rates, and ATP production, and decreased glucose uptake, lactate production, and extracellular acidification rates. For Il33-overexpressing BMDMs, the metabolic reprogramming upon LPS stimulation was similar to WT BMDMs, and was accompanied by increased M1 macrophage activity. Our findings suggested that the pleiotropic IL-33/ST2 pathway may influence the polarization and function of macrophages by regulating mitochondrial metabolism.
Collapse
Affiliation(s)
- Huadan Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yichun He
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiaofeng Yuan
- Department of Pediatrics, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Junqi Niu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dong Li
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China.,Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
40
|
Li SS, Ma J, Wong AST. Chemoresistance in ovarian cancer: exploiting cancer stem cell metabolism. J Gynecol Oncol 2019; 29:e32. [PMID: 29468856 PMCID: PMC5823988 DOI: 10.3802/jgo.2018.29.e32] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is most deadly gynecologic malignancies worldwide. Chemotherapy is the mainstay treatment for ovarian cancer. Despite the initial response is promising, frequent recurrence in patients with advanced diseases remains a therapeutic challenge. Thus, understanding the biology of chemoresistance is of great importance to overcome this challenge and will conceivably benefit the survival of ovarian cancer patients. Although mechanisms underlying the development of chemoresistance are still ambiguous, accumulating evidence has supported an integral role of cancer stem cells (CSCs) in recurrence following chemotherapy. Recently, tumor metabolism has gained interest as a reason of chemoresistance in tumors and chemotherapeutic drugs in combination with metabolism targeting approaches has been found promising in overcoming therapeutic resistance. In this review, we will summarize recent studies on CSCs and metabolism in ovarian cancer and discuss possible role of CSCs metabolism in chemoresistance.
Collapse
Affiliation(s)
- Shan Shan Li
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Jing Ma
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Alice S T Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
41
|
Kobayashi Y, Banno K, Kunitomi H, Takahashi T, Takeda T, Nakamura K, Tsuji K, Tominaga E, Aoki D. Warburg effect in Gynecologic cancers. J Obstet Gynaecol Res 2018; 45:542-548. [PMID: 30511455 DOI: 10.1111/jog.13867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 12/27/2022]
Abstract
Mammalian cells produce energy by oxidative phosphorylation under aerobic conditions. However, in the 1920s, Otto Warburg reported the so-called "Warburg effect" in which cancer cells produce ATP that is biased toward glycolysis rather than mitochondrial oxidative phosphorylation not only in anaerobic environment but also in aerobic environment. Glucose is converted into lactate without going into mitochondria after being metabolized in glycolysis. Compared with oxidative phosphorylation, the glycolysis has a faster ATP production rate but it is very inefficient, resulting in cancer cells consuming a large amount of glucose. Increased glucose metabolism has become a biomarker for cancer cells and has led to the development of positron emission tomography with fluorodeoxyglucose. Till date, the Warburg effect has been an inefficient system for cancer cells with regard to efficient energy production, but since the consumption of oxygen can be suppressed as the tumor grows in mass, it is thought that the Warburg effect is advantageous in this situation wherein the tumor can increase despite the lack of vessels. In addition, an increased lactate by the glycolysis causes acidosis in the microenvironment of tissues, which is thought to damage the surrounding normal tissues and favor the invasion and metastasis of cancer. Thus, Warburg effect is one of the key mechanisms for cancer development and will be the next promising target. In this review, we introduce key players that can be targeted in the Warburg effect and outline the prospects of treatment, targeting the Warburg effect in gynecological cancer.
Collapse
Affiliation(s)
- Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Haruko Kunitomi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Takayuki Takahashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Takeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kanako Nakamura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kosuke Tsuji
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Zhang M, Cong Q, Zhang X, Zhang M, Lu Y, Xu C. Pyruvate dehydrogenase kinase 1 contributes to cisplatin resistance of ovarian cancer through EGFR activation. J Cell Physiol 2018; 234:6361-6370. [PMID: 30229902 DOI: 10.1002/jcp.27369] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Meng Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Qing Cong
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Xiao‐Yan Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai China
| | - Ming‐Xing Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Ying‐Ying Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
| | - Cong‐Jian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University Shanghai China
- Department of Obstetrics and Gynecology of Shanghai Medical School Fudan University Shanghai China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai China
| |
Collapse
|
43
|
Ma Y, Xu Y, Li L. SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling. Exp Ther Med 2018; 16:3195-3201. [PMID: 30233672 DOI: 10.3892/etm.2018.6575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/04/2018] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy worldwide and is one of the five leading causes of cancer-associated mortality in women. There is an urgent requirement to obtain a greater understanding of the molecular mechanism underlying ovarian cancer progression in order to identify novel drug targets and biomarkers. Secreted protein acidic and rich in cysteine-like protein 1 (SPARCL1) has been suggested as a candidate tumor suppressor in various types of human cancers. However, the potential role of SPARCL1 for ovarian cancer has not yet been clearly established. In the present study, lower protein expression levels of SPARCL1 were detected in ovarian cancer tissues when compared with adjacent normal tissues. Overexpression of SPARCL1 significantly suppressed the proliferation and migration of cells from the ovarian cancer cell line SKOV-3, whereas knockdown of SPARCL1 significantly increased cell growth and migration. Furthermore, the results revealed that SPARCL1 overexpression significantly suppressed the activation of the mitogen-activated protein kinase kinase (MEK)/extracellular signal-related kinase (ERK) signaling pathway. Collectively, these results indicated that SPARCL1 may suppress the proliferation and migration of ovarian cancer cells by downregulating signaling via the MEK/ERK pathway.
Collapse
Affiliation(s)
- Yan Ma
- Department of Gynecology, Third Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yuan Xu
- Department of Gynecology, Third Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Li Li
- Department of Gynecology, Third Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
44
|
Zhao XZ, Wu XH. A small compound spindlactone A sensitizes human endometrial cancer cells to TRAIL-induced apoptosis via the inhibition of NAD(P)H dehydrogenase quinone 1. Onco Targets Ther 2018; 11:3609-3617. [PMID: 29950865 PMCID: PMC6016593 DOI: 10.2147/ott.s165723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Introduction Spindlactone A (SPL-A) is a novel small molecule inhibitor of TACC3 that selectively
inhibits the nucleation of centrosome microtubules and induces mitotic arrest in ovarian
cancer cells. SPL-A is derived from dicoumarol which inhibits the activity of NAD(P)H
dehydrogenase quinone oxidoreductase 1 (NQO1). This study aimed to investigate the
mechanism by which SPL-A enhances TRAIL-induced apoptosis in endometrial carcinoma
cells. Materials and methods Endometrial carcinoma cells were treated with SPL-A and/or TRAIL, and the apoptosis and
protein expression in the treated cells were examined. Results Combined treatment with SPL-A and TRAIL significantly induced apoptosis in various
human endometrial carcinoma cells, but not in normal human endometrial stromal cells and
endometrial epithelial cells. Notably, both NQO1 inhibitor ES936 and NQO1 siRNA enhanced
TRAIL-induced apoptosis of endometrial carcinoma cells. Furthermore, SPL-A downregulated
the expression of c-FLIP, Bcl-2, Bcl-xl, and Mcl-1, while increasing p53 expression. Conclusion In particular, luciferase assay showed that SPL-A inhibited Bcl-2 promoter activity,
and p53 inhibitor PFT-α could reverse the effect of SPL-A on Bcl-2 expression.
Moreover, Bcl-2 overexpression inhibited the apoptosis induced by SPL-A and TRAIL. Taken
together, our results suggest that SPL-A sensitizes endometrial cancer cells to
TRAIL-induced apoptosis via the regulation of apoptosis-related proteins and the
inhibition of NQO1 activity.
Collapse
Affiliation(s)
- Xiang-Zhai Zhao
- Department of Gynecology and Obstetrics, The Third Hospital of Hebei Medical University, Hebei 050051, People's Republic of China
| | - Xiao-Hua Wu
- Department of Gynecology and Obstetrics, Hebei Medical University, Hebei 050017, People’s Republic of China.,Department of Gynecology and Obstetrics, Shijiazhuang Obstetrics and Gynecology Hospital, Hebei Medical University, Hebei 050000, People's Republic of China
| |
Collapse
|