1
|
Ichikawa K, Johnson HM, Curtis MA, Biswas N, Singh S, Khachatryan HN, Gater AE, Lin SX, Sperry J. Targeting glioma with heteroaromatic alkaloids: A review of potential therapeutics. Bioorg Med Chem 2025; 121:118051. [PMID: 39999647 DOI: 10.1016/j.bmc.2024.118051] [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: 09/20/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 02/27/2025]
Abstract
Glioblastoma multiforme (GBM), classified as a grade IV astrocytoma, is the most aggressive and deadly form of glioma, characterized by rapid progression, extensive genetic heterogeneity, and resistance to conventional therapies. Despite advancements in surgical techniques, radiation therapy, and the frontline chemotherapeutic agent temozolomide, the prognosis for GBM patients remains poor, with a median survival of 15 months and a 5-year survival rate of approximately 7 %. The absence of effective long-term treatments underscores the urgent, unmet clinical need for novel therapeutic strategies to improve patient outcomes. Natural products, particularly alkaloids, have garnered attention as a rich source of bioactive compounds with diverse pharmacological properties. Alkaloids, a structurally diverse group of natural products, are renowned for their chemotherapeutic properties and ability to cross the blood-brain barrier (BBB), making them promising candidates for glioma therapy. This review systematically examines all reported heteroaromatic alkaloids with documented anti-glioma activities, highlighting their mechanisms of action where available. By providing a comprehensive resource, it aims to facilitate the identification and optimisation of alkaloid-based compounds for glioma-targeted drug discovery. Additionally, this review emphasizes the importance of incorporating natural products into the drug development pipeline to address the pressing challenges associated with glioma, particularly GBM treatment.
Collapse
Affiliation(s)
- Karen Ichikawa
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Hannah M Johnson
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nandita Biswas
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Hasmik N Khachatryan
- Scientific Technological Centre of Organic and Pharmaceutical Chemistry, National Academy of Science of Armenia, Yerevan 0014, Armenia
| | - Anastasia E Gater
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Simon X Lin
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Jonathan Sperry
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand.
| |
Collapse
|
2
|
Reus P, Torbica E, Rothenburger T, Bechtel M, Kandler J, Ciesek S, Gribbon P, Kannt A, Cinatl J, Bojkova D. Papaverine Targets STAT Signaling: A Dual-Action Therapy Option Against SARS-CoV-2. J Med Virol 2025; 97:e70319. [PMID: 40171981 PMCID: PMC11963225 DOI: 10.1002/jmv.70319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 04/04/2025]
Abstract
Papaverine (PV) has been previously identified as a promising candidate in SARS-CoV-2 repurposing screens. In this study, we further investigated both its antiviral and immunomodulatory properties. PV displayed antiviral efficacy against SARS-CoV-2 and influenza A viruses H1N1 and H5N1 in single infection as well as in co-infection. We demonstrated PV's activity against various SARS-CoV-2 variants and identified its action at the post-entry stage of the viral life cycle. Notably, treatment of air-liquid interface (ALI) cultures of primary bronchial epithelial cells with PV significantly inhibited SARS-CoV-2 levels. Additionally, PV was found to attenuate interferon (IFN) signaling independently of viral infection. Mechanistically, PV decreased the activation of the IFN-stimulated response element following stimulation with all three IFN types by suppressing STAT1 and STAT2 phosphorylation and nuclear translocation. Furthermore, the combination of PV with approved COVID-19 therapeutics molnupiravir and remdesivir demonstrated synergistic effects. Given its immunomodulatory effects and clinical availability, PV shows promising potential as a component for combination therapy against COVID-19.
Collapse
Affiliation(s)
- Philipp Reus
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPortHamburgGermany
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Emma Torbica
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Tamara Rothenburger
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Marco Bechtel
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Joshua Kandler
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFrankfurtGermany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPortHamburgGermany
| | - Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPFrankfurtGermany
| | - Jindrich Cinatl
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
- Dr. Petra Joh Research InstituteFrankfurtGermany
| | - Denisa Bojkova
- Institute for Medical Virology, University Hospital, Goethe University FrankfurtFrankfurtGermany
| |
Collapse
|
3
|
Singh IP, Tanwar AK, Chatterjee D, Lal UR. Medicinal chemistry and detoxification of poisonous plants in traditional medicine: A comprehensive review. Toxicon 2025; 258:108307. [PMID: 40056954 DOI: 10.1016/j.toxicon.2025.108307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Poisonous and toxic plants are used in different traditional systems of medicine for medicinal preparations after certain processing (detoxification) method. However, the correlation between plant toxicity and their chemical constituents remains unexplored for many of these plants. A thorough study on bioactive constituents from poisonous plants could also benefit their proper utilization and improve their usage systematically. METHODS A thorough search of research articles and data bases (Science Direct, PubMed, SciFinder, and Google Scholar) have been done for phytoconstituents and their toxicity aspects. Emphasis was upon searching chemistry of detoxification aspects of traditional poisonous plant materials. RESULTS Exploration and correlation with traditional system of medicine would help for future drug research and development. The detoxification methods of poisonous plants have been reported but the chemical changes occurring after detoxification remains unexplored for majority of these plants. To understand the safe and informed usage of these poisonous medicinal plants, it is important to investigate their chemical constituents, toxicity phenotypes, mode of action, underlying mechanisms, and detoxification processing techniques. In the present review, poisonous plants of traditional systems of medicine with their constituents and toxicity have been compiled. CONCLUSION We discussed poisonous medicinal plants and their toxicity, ways to lessen their toxicity, and discover the processes that underlie detoxification and process of detoxification, and the chemical modification of constituents as a result of detoxification. These provide information on how to employ poisonous medicinal plants safely and sensibly in clinics, how to control the quality of toxic herbs. Limited references were available for Papaver somniferum and Cannabis sativa for their restricted (narcotics) use. This review will give valuable insights for researchers in traditional medicine.
Collapse
Affiliation(s)
- Inder Pal Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| | - Ankur Kumar Tanwar
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Debanjan Chatterjee
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Uma Ranjan Lal
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, 160062, Punjab, India
| |
Collapse
|
4
|
Ożarowski M, Karpiński TM, Czerny B, Kamiński A, Seremak-Mrozikiewicz A. Plant Alkaloids as Promising Anticancer Compounds with Blood-Brain Barrier Penetration in the Treatment of Glioblastoma: In Vitro and In Vivo Models. Molecules 2025; 30:1561. [PMID: 40286187 PMCID: PMC11990316 DOI: 10.3390/molecules30071561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Glioblastoma (GBM) is one of the most invasive central nervous system tumors, with rising global incidence. Therapy resistance and poor prognosis highlight the urgent need for new anticancer drugs. Plant alkaloids, a largely unexplored yet promising class of compounds, have previously contributed to oncology treatments. While past reviews provided selective insights, this review aims to collectively compare data from the last decade on (1) plant alkaloid-based anticancer drugs, (2) alkaloid transport across the blood-brain barrier (BBB) in vitro and in vivo, (3) alkaloid mechanisms of action in glioblastoma models (in vitro, in vivo, ex vivo, and in silico), and (4) cytotoxicity and safety profiles. Additionally, innovative drug delivery systems (e.g., nanoparticles and liposomes) are discussed. Focusing on preclinical studies of single plant alkaloids, this review includes 22 botanical families and 28 alkaloids that demonstrated anti-GBM activity. Most alkaloids act in a concentration-dependent manner by (1) reducing glioma cell viability, (2) suppressing proliferation, (3) inhibiting migration and invasion, (4) inducing cell death, (5) downregulating Bcl-2 and key signaling pathways, (6) exhibiting antiangiogenic effects, (7) reducing tumor weight, and (8) improving survival rates. The toxic and adverse effect analysis suggests that alkaloids such as noscapine, lycorine, capsaicin, chelerythrine, caffeine, boldine, and colchicine show favorable therapeutic potential. However, tetrandrine, nitidine, harmine, harmaline, cyclopamine, cocaine, and brucine may pose greater risks than benefits. Piperine's toxicity and berberine's poor bioavailability suggest the need for novel drug formulations. Several alkaloids (kukoamine A, cyclovirobuxine D, α-solanine, oxymatrine, rutaecarpine, and evodiamine) require further pharmacological and toxicological evaluation. Overall, while plant alkaloids show promise in glioblastoma therapy, progress in assessing their BBB penetration remains limited. More comprehensive studies integrating glioma research and advanced drug delivery technologies are needed.
Collapse
Affiliation(s)
- Marcin Ożarowski
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Tomasz M. Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Rokietnicka 10, 60-806 Poznań, Poland
| | - Bogusław Czerny
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Żołnierska 48, 70-204 Szczecin, Poland;
- Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Adam Kamiński
- Department of Orthopaedics and Traumatology, Independent Public Clinical Hospital No. 1, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Agnieszka Seremak-Mrozikiewicz
- Division of Perinatology and Women’s Disease, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland;
- Laboratory of Molecular Biology in Division of Perinatology and Women’s Diseases, University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| |
Collapse
|
5
|
Contreras-Chávez GG, Zapi-Colin LA, Estrada JA, Contreras I, Estrada JA. Advances on the therapeutic potential of cell receptor activation in glioblastoma. Mol Biol Rep 2025; 52:207. [PMID: 39907852 DOI: 10.1007/s11033-025-10312-w] [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: 10/25/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Glioblastoma multiforme is the most common and aggressive malignant brain tumor. Current therapies have been unable to improve life expectancy in patients. This cancer is frequently accompanied by overexpression of receptors, such as EGFR, VEGFR and TLRs, involved in the regulation of inflammation, cell proliferation, differentiation, and survival. The present review summarizes current knowledge from preclinical and clinical studies investigating the role of pattern recognition and tyrosine kinase receptors in glioblastoma development and evolution, and their possible use to improve treatment outcomes and patient survival.
Collapse
Affiliation(s)
- Gerson G Contreras-Chávez
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, México
| | - Luis A Zapi-Colin
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, México
| | - José A Estrada
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, México
| | - Irazú Contreras
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, México.
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza s/n, Colonia Moderna de la Cruz, Toluca, C.P. 50180, México.
| | - José A Estrada
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza s/n, Colonia Moderna de la Cruz, Toluca, C.P. 50180, México
| |
Collapse
|
6
|
Aliebrahimi S, Farnoudian-Habibi A, Heidari F, Amani A, Montazeri V, Sabz Andam S, Saber R, Alizadeh AM, Ostad SN. Using chitosan-coated magnetite nanoparticles as a drug carrier for opioid delivery against breast cancer. Pharm Dev Technol 2024; 29:596-603. [PMID: 38932720 DOI: 10.1080/10837450.2024.2372568] [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: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Over the past decades, opium derivatives have been discovered as new anticancer agents. In our study, Fe3O4 superparamagnetic nanoparticles (SPIONs) decorated with chitosan were loaded with papaverine or noscapine to surmount drug delivery-related obstacles. Modifying the magnetic nanoparticles (MNP) surface with polymeric materials such as chitosan prevents oxidation and provides a site for drug linkage, which renders them a great drug carrier. The obtained systems were characterized by DLS (20-40 nm were achieved for MNPs and drug- loaded MNPs), TEM (spherical with average size of 11-20 nm) FTIR, XRD, and VSM (71.3 - 42.8 emu/g). Contrary to noscapine, papaverine-MNPs attenuated 4T1 murine breast cancer cell proliferation (11.50 ± 1.74 µg/mL) effectively compared to the free drug (62.35 ± 2.88 µg/mL) while sparing L-929 fibroblast cells (138.14 ± 4.38 µg/mL). Furthermore, SPION and SPION-chitosan displayed no cytotoxic activity. Colony-formation assay confirmed the long-term cytotoxicity of nanostructures. Both developed formulations promoted ROS production accompanied by late apoptotic cell death. The biocompatible nanoparticle exerted an augmenting effect to deliver papaverine to metastatic breast cancer cells.
Collapse
Affiliation(s)
- Shima Aliebrahimi
- Department of Artificial Intelligence, Smart University of Medical Sciences, Tehran, Iran
| | - Amir Farnoudian-Habibi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Heidari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Amani
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Vahideh Montazeri
- Department of Artificial Intelligence, Smart University of Medical Sciences, Tehran, Iran
| | - Shiva Sabz Andam
- Department of Medical Nanotechnology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Reza Saber
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Nasser Ostad
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Pasdaran A, Grice ID, Hamedi A. A review of natural products and small-molecule therapeutics acting on central nervous system malignancies: Approaches for drug development, targeting pathways, clinical trials, and challenges. Drug Dev Res 2024; 85:e22180. [PMID: 38680103 DOI: 10.1002/ddr.22180] [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: 05/26/2023] [Revised: 08/09/2023] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
In 2021, the World Health Organization released the fifth edition of the central nervous system (CNS) tumor classification. This classification uses histopathology and molecular pathogenesis to group tumors into more biologically and molecularly defined entities. The prognosis of brain cancer, particularly malignant tumors, has remained poor worldwide, approximately 308,102 new cases of brain and other CNS tumors were diagnosed in the year 2020, with an estimated 251,329 deaths. The cost and time-consuming nature of studies to find new anticancer agents makes it necessary to have well-designed studies. In the present study, the pathways that can be targeted for drug development are discussed in detail. Some of the important cellular origins, signaling, and pathways involved in the efficacy of bioactive molecules against CNS tumorigenesis or progression, as well as prognosis and common approaches for treatment of different types of brain tumors, are reviewed. Moreover, different study tools, including cell lines, in vitro, in vivo, and clinical trial challenges, are discussed. In addition, in this article, natural products as one of the most important sources for finding new chemotherapeutics were reviewed and over 700 reported molecules with efficacy against CNS cancer cells are gathered and classified according to their structure. Based on the clinical trials that have been registered, very few of these natural or semi-synthetic derivatives have been studied in humans. The review can help researchers understand the involved mechanisms and design new goal-oriented studies for drug development against CNS malignancies.
Collapse
Affiliation(s)
- Ardalan Pasdaran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacognosy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Irwin Darren Grice
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, Australia
- School of Medical Science, Griffith University, Gold Coast, Southport, Queensland, Australia
| | - Azadeh Hamedi
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacognosy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Shahcheraghi SH, Alimardani M, Lotfi M, Lotfi M, Uversky VN, Guetchueng ST, Palakurthi SS, Charbe NB, Hromić-Jahjefendić A, Aljabali AAA, Gadewar MM, Malik S, Goyal R, El-Tanani M, Mishra V, Mishra Y, Tambuwala MM. Advances in glioblastoma multiforme: Integrating therapy and pathology perspectives. Pathol Res Pract 2024; 257:155285. [PMID: 38653089 DOI: 10.1016/j.prp.2024.155285] [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: 02/15/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Abstract
Glioblastoma, a highly lethal form of brain cancer, is characterized by its aggressive growth and resistance to conventional treatments, often resulting in limited survival. The response to therapy is notably influenced by various patient-specific genetic factors, underscoring the disease's complexity. Despite the utilization of diverse treatment modalities such as surgery, radiation, and chemotherapy, many patients experience local relapse, emphasizing the critical need for improved therapeutic strategies to effectively target these formidable tumors. Recent years have witnessed a surge in interest in natural products derived from plants, particularly alkaloids, for their potential anticancer effects. Alkaloids have shown promise in cancer chemotherapy by selectively targeting crucial signaling pathways implicated in tumor progression and survival. Specifically, they modulate the NF-κB and MAPK pathways, resulting in reduced tumor growth and altered gene expression across various cancer types. Additionally, alkaloids exhibit the capacity to induce cell cycle arrest, further impeding tumor proliferation in several malignancies. This review aims to delineate recent advances in understanding the pathology of glioblastoma multiforme (GBM) and to explore the potential therapeutic implications of alkaloids in managing this deadly disease. By segregating discussions on GBM pathology from those on alkaloid-based therapies, we provide a structured overview of the current challenges in GBM treatment and the promising opportunities presented by alkaloid-based interventions. Furthermore, we briefly discuss potential future directions in GBM research and therapy beyond alkaloids, including emerging treatment modalities or areas of investigation that hold promise for improving patient outcomes. In conclusion, our efforts offer hope for enhanced outcomes and improved quality of life for GBM patients through alkaloid-based therapies. By integrating insights from pathology and therapeutic perspectives, we underscore the significance of a comprehensive approach in addressing this devastating disease.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maliheh Alimardani
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Lotfi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Stephanie Tamdem Guetchueng
- Institute of Medical Research and Medicinal Plants Studies, Ministry of Scientific Research and Innovation, PO Box 6163, Yaoundé, Cameroon
| | - Sushesh Shrivastsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School Of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Nitin B Charbe
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Manoj M Gadewar
- Department of Pharmacology, School of medical and allied sciences, K.R. Mangalam University, Gurgaon, Haryana 122103, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan, India
| | - Mohamed El-Tanani
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Murtaza M Tambuwala
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln LN6 7TS, UK.
| |
Collapse
|
9
|
Kurasaka C, Nishizawa N, Ogino Y, Sato A. Anticancer sensitivity and biological aspect of 5-fluorouracil-resistant human colorectal cancer HCT116 cells in three-dimensional culture under high- and low-glucose conditions. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:870-880. [PMID: 38555594 DOI: 10.1080/15257770.2024.2332414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
5-Fluorouracil (5-FU) is a commonly used anticancer drug for colorectal cancer (CRC). Therefore, it is crucial to elucidate the mechanisms that contribute to 5-FU resistance. We established an acquired 5-FU resistant cell line, HCT116RF10, derived from CRC cells and investigated its energy metabolism as well as the underlying mechanism of 5-FU resistance. We examined the sensitivity to 5-FU and the formation of tumor spheres in parental HCT116 cells and 5-FU-resistant HCT116RF10 cells under 3D culture conditions at high-glucose (HG 25 mM) and low-glucose (LG 5.5 mM) concentrations. These results suggested that the tumor spheres of parental HCT116 cells displayed higher sensitivity to 5-FU under LG conditions than under HG conditions. HCT116RF10 tumor spheres exhibited comparable sensitivity to 5-FU under HG and LG conditions. Furthermore, under HG conditions, there was a marked decrease in extracellular lactate in the HCT116RF10 tumor sphere compared to that in the LG tumor sphere. Similarly, HCT116 tumor spheres showed decreased extracellular lactate levels under LG conditions compared to those grown under HG conditions. Moreover, the evidence reveals that the tumor spheres of HCT116RF10 and HCT116 cells exhibit disparate dependencies on energy metabolism, glycolysis, and mitochondrial respiration under both HG and LG conditions. These results have important clinical implications for overcoming 5-FU resistance and enhancing antitumor treatment strategies.
Collapse
Affiliation(s)
- Chinatsu Kurasaka
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Nana Nishizawa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yoko Ogino
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Akira Sato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
10
|
Cao J, Wang H, Cao Y, Kan S, Li J, Liu Y. Extreme Reconfiguration of Plastid Genomes in Papaveraceae: Rearrangements, Gene Loss, Pseudogenization, IR Expansion, and Repeats. Int J Mol Sci 2024; 25:2278. [PMID: 38396955 PMCID: PMC10888665 DOI: 10.3390/ijms25042278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
The plastid genomes (plastomes) of angiosperms are typically highly conserved, with extreme reconfiguration being uncommon, although reports of such events have emerged in some lineages. In this study, we conducted a comprehensive comparison of the complete plastomes from twenty-two species, covering seventeen genera from three subfamilies (Fumarioideae, Hypecooideae, and Papaveroideae) of Papaveraceae. Our results revealed a high level of variability in the plastid genome size of Papaveraceae, ranging from 151,864 bp to 219,144 bp in length, which might be triggered by the expansion of the IR region and a large number of repeat sequences. Moreover, we detected numerous large-scale rearrangements, primarily occurring in the plastomes of Fumarioideae and Hypecooideae. Frequent gene loss or pseudogenization were also observed for ndhs, accD, clpP, infA, rpl2, rpl20, rpl32, rps16, and several tRNA genes, particularly in Fumarioideae and Hypecooideae, which might be associated with the structural variation in their plastomes. Furthermore, we found that the plastomes of Fumarioideae exhibited a higher GC content and more repeat sequences than those of Papaveroideae. Our results showed that Papaveroideae generally displayed a relatively conserved plastome, with the exception of Eomecon chionantha, while Fumarioideae and Hypecooideae typically harbored highly reconfigurable plastomes, showing high variability in the genome size, gene content, and gene order. This study provides insights into the plastome evolution of Papaveraceae and may contribute to the development of effective molecular markers.
Collapse
Affiliation(s)
- Jialiang Cao
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (J.C.); (H.W.); (Y.C.)
| | - Hongwei Wang
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (J.C.); (H.W.); (Y.C.)
| | - Yanan Cao
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (J.C.); (H.W.); (Y.C.)
| | - Shenglong Kan
- Marine College, Shandong University, Weihai 264209, China;
| | - Jiamei Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450046, China
| | - Yanyan Liu
- College of Plant Protection, Henan Agricultural University, Zhengzhou 450002, China; (J.C.); (H.W.); (Y.C.)
| |
Collapse
|
11
|
Tsai WE, Liu YT, Kuo FH, Cheng WY, Shen CC, Chiao MT, Huang YF, Liang YJ, Yang YC, Hsieh WY, Chen JP, Liu SY, Chiu CD. Crocetin Enhances Temozolomide Efficacy in Glioblastoma Therapy Through Multiple Pathway Suppression. Curr Neurovasc Res 2024; 21:320-336. [PMID: 39092730 DOI: 10.2174/0115672026332275240731054001] [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: 05/23/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive type of brain tumor that is difficult to remove surgically. Research suggests that substances from saffron, namely crocetin and crocin, could be effective natural treatments, showing abilities to kill cancer cells. METHODS Our study focused on evaluating the effects of crocetin on glioma using the U87 cell line. We specifically investigated how crocetin affects the survival, growth, and spread of glioma cells, exploring its impact at concentrations ranging from 75-150 μM. The study also included experiments combining crocetin with the chemotherapy drug Temozolomide (TMZ) to assess potential synergistic effects. RESULTS Crocetin significantly reduced the viability, proliferation, and migration of glioma cells. It achieved these effects by decreasing the levels of Matrix Metallopeptidase 9 (MMP-9) and Ras homolog family member A (RhoA), proteins that are critical for cancer progression. Additionally, crocetin inhibited the formation of cellular structures necessary for tumor growth. It blocked multiple points of the Ak Strain Transforming (AKT) signaling pathway, which is vital for cancer cell survival. This treatment led to increased cell death and disrupted the cell cycle in the glioma cells. When used in combination with TMZ, crocetin not only enhanced the reduction of cancer cell growth but also promoted cell death and reduced cell replication. This combination therapy further decreased levels of high mobility group box 1 (HMGB1) and Receptor for Advanced Glycation End-products (RAGE), proteins linked to inflammation and tumor progression. It selectively inhibited certain pathways involved in the cellular stress response without affecting others. CONCLUSION Our results underscore the potential of crocetin as a treatment for glioma. It targets various mechanisms involved in tumor growth and spread, offering multiple avenues for therapy. Further studies are essential to fully understand and utilize crocetin's benefits in treating glioma.
Collapse
Affiliation(s)
- Wei-En Tsai
- Taichung Municipal Taichung First Senior High School, Taichung, Taiwan
| | - Yen-Tsen Liu
- Taichung Municipal Taichung First Senior High School, Taichung, Taiwan
| | - Fu-Hsuan Kuo
- Center for Geriatrics and Gerontology, Taichung Veterans Hospital, Taichung, 40705, Taiwan
| | - Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chiung-Chyi Shen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan
- Basic Medical Education, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Ming-Tsang Chiao
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Fen Huang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yea-Jiuen Liang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Chin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wan-Yu Hsieh
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jun-Peng Chen
- Biostatistics Task Force, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Szu-Yuan Liu
- Department of Neurosurgery, Oncology Neurosurgery Division, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Science, College of Life Science, Graduate Institute of Life Science, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Di Chiu
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
12
|
Akram S, Al-Shammari AM, Sahib HB, Jabir MS. Papaverine Enhances the Oncolytic Effects of Newcastle Disease Virus on Breast Cancer In Vitro and In Vivo. Int J Microbiol 2023; 2023:3324247. [PMID: 37720338 PMCID: PMC10504052 DOI: 10.1155/2023/3324247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/28/2022] [Accepted: 08/20/2023] [Indexed: 09/19/2023] Open
Abstract
Breast cancer is a lethal disease in females worldwide and needs effective treatment. Targeting cancer cells with selective and safe treatment seems like the best choice, as most chemotherapeutic drugs act unselectively. Papaverine showed promising antitumor activity with a high safety profile and increased blood flow through vasodilation. At the same time, it was widely noticed that virotherapy using the Newcastle disease virus proved to be safe and selective against a broad range of cancer cells. Furthermore, combination therapy is favorable, as it attacks cancer cells with multiple mechanisms and enhances virus entrance into the tumor mass, overcoming cancer cells' resistance to therapy. Therefore, we aimed at assessing the novel combination of the AMHA1 strain of Newcastle disease virus (NDV) and nonnarcotic opium alkaloid (papaverine) against breast cancer models in vitro and in vivo. Methods. In vitro experiments used two human breast cancer cell lines and one normal cell line and were treated with NDV, papaverine, and a combination. The study included a cell viability MTT assay, morphological analysis, and apoptosis detection. Animal experiments used the AN3 mouse mammary adenocarcinoma tumor model. Evaluation of the antitumor activity included growth inhibition measurement; the immunohistochemistry assay measured caspase protein expression. Finally, a semiquantitative microarray assay was used to screen changes in apoptotic proteins. In vitro, results showed that the combination therapy induces synergistic cytotoxicity and apoptosis against cancer cells with a negligible cytotoxic effect on normal cells. In vivo, combination treatment induced a significant antitumor effect with an obvious regression in tumor size and a remarkable and significant expression of caspase-3, caspase-8, and caspase-9 compared to monotherapies. Microarray analysis shows higher apoptosis protein levels in the combination therapy group. In conclusion, this study demonstrated the role of papaverine in enhancing the antitumor activity of NDV, suggesting a promising strategy for breast cancer therapy through nonchemotherapeutic drugs.
Collapse
Affiliation(s)
- Sura Akram
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Ahmed Majeed Al-Shammari
- Experimental Therapy, Iraqi Center for Cancer and Medical Genetics Research, Mustansiriyah University, Baghdad, Iraq
| | - Hayder B. Sahib
- Department of Pharmacology, College of Pharmacy, Al-Nahrain University, Baghdad, Iraq
| | - Majid Sakhi Jabir
- Department of Applied Science, University of Technology, Baghdad, Iraq
| |
Collapse
|
13
|
Özkul B, Sever İH, Yiğittürk G, Elgörmüş ÇS, Gür SG, Erbaş O. Demonstration of ameliorating effect of papaverine in sepsis-induced acute lung injury on rat model through radiology and histology. ULUS TRAVMA ACIL CER 2023; 29:963-971. [PMID: 37681716 PMCID: PMC10560817 DOI: 10.14744/tjtes.2023.73580] [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: 07/24/2022] [Revised: 07/26/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Our target was to show the role of high mobility group box-1/receptor for (HMGB1/RAGE) interaction in feces intraperitoneal injection procedure (FIP)-induced acute lung injury (ALI) pathophysiology, to investigate the effect of papaverine on RAGE associated NF-κB pathway by determining the level of soluble RAGE (sRAGE) and HMGB1, and to support this hypothesis by evaluating inflammatory biochemical, oxidative stress markers, Hounsfield unit (HU) value in computed tomography (CT), and histo-pathological results. METHODS FIP was performed on 37 Wistar rats for creating a sepsis-induced ALI model. The animals were assigned into four groups as follows: Normal control (no treatment), placebo (FIP and saline), and receiving 20 mg/kg and 40 mg/kg per day papaverine. Twenty h after FIP, CT examination was performed for all animals, and HU value of the lung parenchyma was measured. The plasma levels of tumor necrosis factor (TNF)-α, HMGB1, sRAGE, C-reactive protein (CRP) and malondialdehyde (MDA), and lactic acid (LA) were determined and PaO2 and PaCO2 were measured from arterial blood sample. Lung damage was assessed by histopathological. RESULTS TNF-, IL-6, CRP, HMGB1, MDA, LA levels, histopathologic scores, and HU values of CT were significantly increased and sRAGE levels were decreased in the saline-treated group against normal group (all P<0.05). Papaverine significantly reversed all results regardless of the dose (all P<0.05) and demonstrated inhibition of HMGB1/RAGE interaction through increasing sRAGE levels and suppresses the pro-inflammatory cytokines. CONCLUSION We concluded that papaverine has ameliorating effects in rat model of ALI.
Collapse
Affiliation(s)
- Bahattin Özkul
- Department of Radiology, İstanbul Atlas University, İstanbul-Türkiye
| | | | - Gürkan Yiğittürk
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Muğla-Türkiye
| | | | | | - Oytun Erbaş
- Department of Physiology, Demiroğlu Bilim University, İstanbul-Türkiye
| |
Collapse
|
14
|
Okamoto R, Toya K, Ogino Y, Sato A. Downregulation of long noncoding RNA TP73-AS1 expression confers resistance to temozolomide in human glioblastoma cells. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:86-98. [PMID: 37452786 DOI: 10.1080/15257770.2023.2234960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Glioblastoma multiforme (GBM), the most aggressive primary malignant brain tumor, is resistant to conventional radiotherapies and chemotherapies, including temozolomide (TMZ). Overcoming GBM resistance to the chemotherapeutic agent TMZ poses an important therapeutic problem. This study established an association between the long noncoding RNA TP73-AS1 and TMZ sensitivity regulation in human GBM cells (U87MG). Transcriptomic analysis revealed that TP73-AS1 expression was reduced in TMZ-resistant U87MGRT100 cells compared to that in parental U87MG cells. Additionally, TP73-AS1 knockdown in parental U87MG cells decreased their sensitivity to TMZ. Overall, these findings suggest that TP73-AS1 functions as a regulator of TMZ sensitivity in GBM cells.
Collapse
Affiliation(s)
- Ryo Okamoto
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Koki Toya
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yoko Ogino
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Akira Sato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
15
|
Ashrafi S, Alam S, Sultana A, Raj A, Emon NU, Richi FT, Sharmin T, Moon M, Park MN, Kim B. Papaverine: A Miraculous Alkaloid from Opium and Its Multimedicinal Application. Molecules 2023; 28:3149. [PMID: 37049912 PMCID: PMC10095881 DOI: 10.3390/molecules28073149] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
The pharmacological actions of benzylisoquinoline alkaloids are quite substantial, and have recently attracted much attention. One of the principle benzylisoquinoline alkaloids has been found in the unripe seed capsules of Papaver somniferum L. Although it lacks analgesic effects and is unrelated to the compounds in the morphine class, it is a peripheral vasodilator and has a direct effect on vessels. It is reported to inhibit the cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) phosphodiesterase in smooth muscles, and it has been observed to increase intracellular levels of cAMP and cGMP. It induces coronary, cerebral, and pulmonary artery dilatation and helps to lower cerebral vascular resistance and enhance cerebral blood flow. Current pharmacological research has revealed that papaverine demonstrates a variety of biological activities, including activity against erectile dysfunction, postoperative vasospasms, and pulmonary vasoconstriction, as well as antiviral, cardioprotective, anti-inflammatory, anticancer, neuroprotective, and gestational actions. It was recently demonstrated that papaverine has the potential to control SARS-CoV-2 by preventing its cytopathic effect. These experiments were carried out both in vitro and in vivo and require an extensive understanding of the mechanisms of action. With its multiple mechanisms, papaverine can be considered as a natural compound that is used to develop therapeutic drugs. To validate its applications, additional research is required into its precise therapeutic mechanisms as well as its acute and chronic toxicities. Therefore, the goal of this review is to discuss the major studies and reported clinical studies looking into the pharmacological effects of papaverine and the mechanisms of action underneath these effects. Additionally, it is recommended to conduct further research via significant pharmacodynamic and pharmacokinetic studies.
Collapse
Affiliation(s)
- Sania Ashrafi
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Safaet Alam
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
- Drugs and Toxins Research Division, BCSIR Laboratories Rajshahi, Bangladesh Council of Scientific and Industrial Research, Rajshahi 6206, Bangladesh
| | - Arifa Sultana
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Asef Raj
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Nazim Uddin Emon
- Department of Pharmacy, Faculty of Science and Engineering, International Islamic University Chittagong, Chittagong 4318, Bangladesh
- Department of Chemistry and Biochemistry, Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Fahmida Tasnim Richi
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Tasnuva Sharmin
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Myunghan Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Sharma S, Kumar P. Decoding the Role of MDM2 as a Potential Ubiquitin E3 Ligase and Identifying the Therapeutic Efficiency of Alkaloids against MDM2 in Combating Glioblastoma. ACS OMEGA 2023; 8:5072-5087. [PMID: 36777618 PMCID: PMC9910072 DOI: 10.1021/acsomega.2c07904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/17/2023] [Indexed: 05/28/2023]
Abstract
Glioblastomas (GBMs) represent the most aggressive form of brain tumor arising from the malignant transformation of astrocytes. Despite various advancements, treatment options remain limited to chemotherapy and radiotherapy followed by surgery giving an overall survival of 14-15 months. These therapies are somewhere restricted in giving a better survival and cure. There is a need for new therapeutics that could potentially target GBM based on molecular pathways and pathology. Here, ubiquitin E3 ligases can be used as targets as they bind a wide array of substrates and therefore can be attractive targets for new inhibitors. Through this study, we have tried to sort various ubiquitin E3 ligases based on their expression, pathways to which these ligases are associated, and mutational frequencies, and then we tried to screen potent inhibitors against the most favorable E3 ligase as very few studies are available concerning inhibition of E3 ligase in GBM. Our study found MDM2 to be the most ideal E3 ligase and further we tried to target MDM2 against various compounds under the alkaloid class. Molecular Docking and MD simulations combined with ADMET properties and BBB scores revealed that only evodiamine and sanguinarine were effective in inhibiting MDM2. We also tried to give a proposed mechanism of how these inhibitors mediate the p53 signaling in GBM. Therefore, the new scaffolds predicted by the computational approach could help in designing promising therapeutic agents targeting MDM2 in glioblastoma.
Collapse
|
17
|
RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int J Mol Sci 2022; 24:ijms24010266. [PMID: 36613714 PMCID: PMC9820344 DOI: 10.3390/ijms24010266] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/28/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin family that is overexpressed in several cancers. RAGE is highly expressed in the lung, and its expression increases proportionally at the site of inflammation. This receptor can bind a variety of ligands, including advanced glycation end products, high mobility group box 1, S100 proteins, adhesion molecules, complement components, advanced lipoxidation end products, lipopolysaccharides, and other molecules that mediate cellular responses related to acute and chronic inflammation. RAGE serves as an important node for the initiation and stimulation of cell stress and growth signaling mechanisms that promote carcinogenesis, tumor propagation, and metastatic potential. In this review, we discuss different aspects of RAGE and its prominent ligands implicated in cancer pathogenesis and describe current findings that provide insights into the significant role played by RAGE in cancer. Cancer development can be hindered by inhibiting the interaction of RAGE with its ligands, and this could provide an effective strategy for cancer treatment.
Collapse
|
18
|
The other side of the coin: Positive view on the role of opioids in cancer. Eur J Pharmacol 2022; 923:174888. [PMID: 35367422 DOI: 10.1016/j.ejphar.2022.174888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 11/23/2022]
Abstract
Opioids have been used for medicinal purposes as an analgesic and recreational purposes as a euphorigenic throughout human history. Cancer patients are often treated with different doses of opioids concurrently with anti-cancer drugs for pain relief without exhibiting excessive adverse effects. The intersection of the biology of pain, opioid therapy, and disease progression represents the crux of the matters and is of potentially great importance in cancer care. For more than 20 years, multiple investigations have focused on the stimulatory effects of opioids on cancer cell growth, while in-depth studies on the inhibitory effects on cancer cell growth development have usually been neglected. This paper reviews the evidence regarding opioid therapies and their anti-cancer effects on various malignancies. Likewise, we have a glimpse into the molecular mechanisms necessary for pinpointing their positive or negative impacts on malignancies to raise awareness and stimulate more excellent dialogue regarding their carcinogenic/anticarcinogenic roles.
Collapse
|
19
|
Gomes DA, Joubert AM, Visagie MH. In Vitro Effects of Papaverine on Cell Migration and Vascular Endothelial Growth Factor in Cancer Cell Lines. Int J Mol Sci 2022; 23:4654. [PMID: 35563045 PMCID: PMC9104338 DOI: 10.3390/ijms23094654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Papaverine (PPV) is a benzylisoquinoline alkaloid isolated from Papaver somniferum that exerts antiproliferative activity. However, several questions remain regarding the biochemical pathways affected by PPV in tumourigenic cells. In this study, the influence of PPV on cell migration (light microscopy), expression of vascular endothelial growth factor (VEGF) B, VEGF R1, VEGF R2, and phosphorylated focal adhesion kinase (pFAK) were investigated using spectrophotometry in MDA-MB-231-, A549- and DU145 cell lines. The migration assay revealed that, after 48 h, PPV (100 µM) reduced cell migration to 81%, 91%, and 71% in MDA-MB-231-, A549-, and DU145 cells, respectively. VEGF B expression was reduced to 0.79-, 0.71-, and 0.73-fold after 48 h of exposure to PPV in MDA-MB-231-, A549- and DU145 cells, while PPV exposure of 48 h increased VEGF R1 expression in MDA-MB-231- and DU145 cells to 1.38 and 1.46. A fold decrease in VEGF R1 expression was observed in A549 cells to 0.90 after exposure to 150 µM. No statistically significant effects were observed on VEGF R2- and FAK expression after exposure to PPV. This study contributes to the understanding of the effects of a phytomedicinal alkaloid compound in cancer cells and may provide novel approaches to the application of non-addictive alkaloids.
Collapse
Affiliation(s)
| | | | - Michelle Helen Visagie
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa; (D.A.G.); (A.M.J.)
| |
Collapse
|
20
|
Vikram ENT, Ilavarasan R, Kamaraj R. Anti-cancer activities of Schedule E1 drugs used in ayurvedic formulations. J Ayurveda Integr Med 2022; 13:100545. [PMID: 35661925 PMCID: PMC9163510 DOI: 10.1016/j.jaim.2022.100545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/13/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022] Open
Abstract
Schedule E1 is an important part of Drugs and Cosmetics Act (Government of India) that comprises the list of poisonous drugs from plant, animal and mineral origins to be consumed under medical supervision. Ayurveda, the world's oldest medicinal system has a list of drugs represented in schedule E1 that are used since thousands of years. This review reports the anti-cancer activities of fifteen toxic ayurvedic drugs from plant origin represented in Drugs and Cosmetics Act, 1940. The information was collected from the various authentic sources, compiled and summarised. The plant extracts, formulations, phytoconstituents and other preparations of these drugs have shown effective activities against mammary carcinoma, neuroblastoma, non-small cell lung carcinoma, lymphocytic leukaemia, colorectal adenocarcinoma, Ehrlich ascites carcinoma, prostate adenocarcinoma, glioblastoma asterocytoma and other malignancies. They have various mechanisms of action including Bax upregulation, Bcl2 downregulation, induction of cell cycle arrest at S phase, G2/M phase, inhibition of vascular endothelial growth factors, inhibition of Akt/mTOR signalling etc. Certain traditional ayurvedic preparations containing these plants are reported beneficial and the possibilities of these drugs as the alternative and adjuvant therapeutic agents in the current cancer care have been discussed. The studies suggest that these drugs could be utilised in future for the critical care of malignancies.
Collapse
Affiliation(s)
- E N T Vikram
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Kancheepuram (Dt.), Tamilnadu 603203, India
| | - R Ilavarasan
- Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Arumbakkam, Chennai, Tamilnadu 600106, India
| | - R Kamaraj
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Kancheepuram (Dt.), Tamilnadu 603203, India.
| |
Collapse
|
21
|
Tanuma SI, Oyama T, Okazawa M, Yamazaki H, Takao K, Sugita Y, Amano S, Abe T, Sakagami H. A Dual Anti-Inflammatory and Anti-Proliferative 3-Styrylchromone Derivative Synergistically Enhances the Anti-Cancer Effects of DNA-Damaging Agents on Colon Cancer Cells by Targeting HMGB1-RAGE-ERK1/2 Signaling. Int J Mol Sci 2022; 23:ijms23073426. [PMID: 35408786 PMCID: PMC8998738 DOI: 10.3390/ijms23073426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 01/27/2023] Open
Abstract
The current anti-cancer treatments are not enough to eradicate tumors, and therefore, new modalities and strategies are still needed. Most tumors generate an inflammatory tumor microenvironment (TME) and maintain the niche for their development. Because of the critical role of inflammation via high-mobility group box 1 (HMGB1)–receptor for advanced glycation end-products (RAGE) signaling pathway in the TME, a novel compound possessing both anti-cancer and anti-inflammatory activities by suppressing the HMGB1-RAGE axis provides an effective strategy for cancer treatment. A recent work of our group found that some anti-cancer 3-styrylchromones have weak anti-inflammatory activities via the suppression of this axis. In this direction, we searched such anti-cancer molecules possessing potent anti-inflammatory activities and discovered 7-methoxy-3-hydroxy-styrylchromone (C6) having dual suppressive activities. Mechanism-of-action studies revealed that C6 inhibited the increased phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) under the stimulation of HMGB1-RAGE signaling and thereby suppressed cytokine production in macrophage-like RAW264.7 cells. On the other hand, in colorectal cancer HCT116 cells, C6 inhibited the activation of ERK1/2, cyclin-dependent kinase 1, and AKT, down-regulated the protein level of XIAP, and up-regulated pro-apoptotic Bax and caspase-3/7 expression. These alterations are suggested to be involved in the C6-induced suppression of cell cycle/proliferation and initiation of apoptosis in the cancer cells. More importantly, in cancer cells, the treatment of C6 potentiates the anti-cancer effects of DNA-damaging agents. Thus, C6 may be a promising lead for the generation of a novel class of cancer therapeutics.
Collapse
Affiliation(s)
- Sei-ichi Tanuma
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda 278-8510, Chiba, Japan; (T.O.); (M.O.); (H.Y.)
- Research Institute of Odontology (M-RIO), School of Dentistry, Meikai University, Sakado 350-0283, Saitama, Japan; (S.A.); (H.S.)
- Correspondence:
| | - Takahiro Oyama
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda 278-8510, Chiba, Japan; (T.O.); (M.O.); (H.Y.)
- Hinoki Shinyaku Co., Ltd., Chiyoda-ku 102-0084, Tokyo, Japan;
| | - Miwa Okazawa
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda 278-8510, Chiba, Japan; (T.O.); (M.O.); (H.Y.)
| | - Hiroaki Yamazaki
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda 278-8510, Chiba, Japan; (T.O.); (M.O.); (H.Y.)
| | - Koichi Takao
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Sakado 350-0295, Saitama, Japan; (K.T.); (Y.S.)
| | - Yoshiaki Sugita
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Sakado 350-0295, Saitama, Japan; (K.T.); (Y.S.)
| | - Shigeru Amano
- Research Institute of Odontology (M-RIO), School of Dentistry, Meikai University, Sakado 350-0283, Saitama, Japan; (S.A.); (H.S.)
| | - Takehiko Abe
- Hinoki Shinyaku Co., Ltd., Chiyoda-ku 102-0084, Tokyo, Japan;
| | - Hiroshi Sakagami
- Research Institute of Odontology (M-RIO), School of Dentistry, Meikai University, Sakado 350-0283, Saitama, Japan; (S.A.); (H.S.)
| |
Collapse
|
22
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Elucidating Antiangiogenic Potential of Rauwolfia serpentina: VEGFR-2 Targeting-Based Molecular Docking Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6224666. [PMID: 35198035 PMCID: PMC8860507 DOI: 10.1155/2022/6224666] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis plays a critical role in tumorigenesis as it provides the necessary blood supply to the newly grown solid tumor. It helps maintain the tumor microenvironment, promotes tumor development, progression, and metastasis. The vascular epithelial growth factor (VEGF), interacting with the tyrosine kinase receptor VEGFR-2 on endothelial cells, exerts its proangiogenic activity. Hence, targeting the VEGFR-2 signaling is considered a promising strategy to inhibit angiogenesis and thus cancer treatment. This study aims to identify the bioactive compounds derived from the medicinal herb Rauwolfia serpentina that effectively binds with VEGFR-2. The bioactive compounds of R. serpentina were first screened for their physicochemical properties using the DataWarrior program (version 5.5.0). Finally, 17 compounds that obeyed Lipinski's rule of five and showed good drug-likeness were selected for molecular docking studies. Molecular docking results showed that the ligands ajmalicidine, 1, 2-dihydrovomilenine, rauwolscine, yohimbine, ajmaline, and papaverine interact strongly with the target VEGFR-2 receptor. Hydrogen bonds and hydrophobic interactions stabilized the interactions of these compounds with VEGFR-2. These compounds showed favourable drug-like properties and possess no significant toxicity. Therefore, the findings of this study indicate that the compounds derived from R. serpentina can be considered for the development of antiangiogenic drug candidates by targeting VEGFR-2.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Butnariu M, Quispe C, Herrera-Bravo J, Pentea M, Sarac I, Küşümler AS, Özçelik B, Painuli S, Semwal P, Imran M, Gondal TA, Emamzadeh-Yazdi S, Lapava N, Yousaf Z, Kumar M, Eid AH, Al-Dhaheri Y, Suleria HAR, del Mar Contreras M, Sharifi-Rad J, Cho WC. Papaver Plants: Current Insights on Phytochemical and Nutritional Composition Along with Biotechnological Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2041769. [PMID: 36824615 PMCID: PMC9943628 DOI: 10.1155/2022/2041769] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/13/2021] [Accepted: 01/06/2022] [Indexed: 11/17/2022]
Abstract
The genus Papaver is highly esteemed in the pharmacy industry, in the culinary field, and as ornamental plants. These plants are also valued in traditional medicine. Among all Papaver species, Papaver somniferum L. (opium poppy) is the most important species in supplying phytochemicals for the formulation of drugs, mainly alkaloids like morphine, codeine, rhoeadine, thebaine, and papaverine. In addition, Papaver plants present other types of phytochemicals, which altogether are responsible for its biological activities. Therefore, this review covers the phytochemical composition of Papaver plants, including alkaloids, phenolic compounds, and essential oils. The traditional uses are reviewed along with their pharmacological activities. Moreover, safety aspects are reported to provide a deep overview of the pharmacology potential of this genus. An updated search was carried out in databases such as Google Scholar, ScienceDirect, and PubMed to retrieve the information. Overall, this genus is a rich source of alkaloids of different types and also contains interesting phenolic compounds, such as anthocyanins, flavonols, and the characteristic indole derivatives nudicaulins. Among other pharmacological properties, numerous preclinical studies have been published about the analgesic, anticancer, antimicrobial, antioxidant, and antidiabetic activities of Papaver plants. Although it highlights the significant impact of this genus for the treatment of a variety of diseases and conditions, as a future prospect, characterization works accompanying preclinical studies are required along with clinical and toxicology studies to establish a correlation between the scientific and traditional knowledge.
Collapse
Affiliation(s)
- Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Timisoara, Romania
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Jesús Herrera-Bravo
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Chile
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
| | - Marius Pentea
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Timisoara, Romania
| | - Ioan Sarac
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Timisoara, Romania
| | - Aylin Seylam Küşümler
- İstanbul Okan University, Nutrition and Dietetics Department, Tuzla, İstanbul, Turkey
| | - Beraat Özçelik
- Department Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, 34469 Istanbul, Turkey
- BIOACTIVE Research & Innovation Food Manufacturing Industry Trade LTD Co., Maslak, Istanbul 34469, Turkey
| | - Sakshi Painuli
- Department of Biotechnology, Graphic Era University, 248001, Dehradun, Uttarakhand, India
- Himalayan Environmental Studies and Conservation Organization, Prem Nagar, Dehradun, 248001 Uttarakhand, India
| | - Prabhakar Semwal
- Department of Biotechnology, Graphic Era University, 248001, Dehradun, Uttarakhand, India
- Department of Life Sciences, Graphic Era Deemed to be University, Dehradun-248002, Uttarakhand, India
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Pakistan
| | | | - Simin Emamzadeh-Yazdi
- Department of Plant and Soil Sciences, University of Pretoria, Gauteng 0002, South Africa
| | - Natallia Lapava
- Medicine Standardization Department of Vitebsk State Medical University, Belarus
| | | | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, 400019, Mumbai, India
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Yusra Al-Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, UAE
| | | | - María del Mar Contreras
- Department of Chemical, Environmental and Materials Engineering, Universidad de Jaén, Campus las Lagunillas, s/n, 23071 Jaén, Spain
| | | | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| |
Collapse
|
24
|
Anju T, Rai NKSR, Kumar A. Sauropus androgynus (L.) Merr.: a multipurpose plant with multiple uses in traditional ethnic culinary and ethnomedicinal preparations. JOURNAL OF ETHNIC FOODS 2022; 9:10. [PMCID: PMC8900104 DOI: 10.1186/s42779-022-00125-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Various plants form the basis of multiple traditional ethnic cuisines and ethnomedicinal practices across the globe. The ethnic cuisines cater to the nutritional, dietary and medicinal requirements of the tribal and rural communities even today. Using literature from various scholarly databases, this study was conducted to consolidate a comprehensive review on the use of Sauropus androgynus (L.) Merr. in various traditional ethnic cuisines and ethnomedicinal preparations across the globe. The survey shows that it is used in multiple ethnic cuisines and is variously known in different countries and among the communities. Further, it possesses multiple nutritional and ethnomedicinal properties. Considering its importance in ethnic foods and ethnomedicinal preparations, it is important to investigate the nutritional composition, phytochemical constitution and pharmacological basis of ethnomedicinal uses. Therefore, we further compiled this information and found that it is a rich source of both micro- and macronutrients and packed with several bioactive compounds. Survey of pharmacological studies on its traditional medicinal uses supports its ethnomedicinal properties. Despite its importance in traditional food and ethnomedicinal systems, it remains underexplored. Limited information on the toxicity of its various extracts shows that further studies should be conducted to understand its safety aspects. Further clinical studies to prospect possible drug candidates from it should be attempted.
Collapse
Affiliation(s)
- Thattantavide Anju
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| | - Nishmitha Kumari S. R. Rai
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| | - Ajay Kumar
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| |
Collapse
|
25
|
Fessner ND, Grimm C, Srdič M, Weber H, Kroutil W, Schwaneberg U, Glieder A. Natural Product Diversification by One‐Step Biocatalysis using Human P450 3A4. ChemCatChem 2021. [DOI: 10.1002/cctc.202101564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nico D. Fessner
- Institute of Molecular Biotechnology NAWI Graz Graz University of Technology Petersgasse 14 8010 Graz Austria
| | - Christopher Grimm
- Institute of Chemistry NAWI Graz University of Graz Heinrichstraße 28 8010 Graz Austria
| | - Matic Srdič
- SeSaM-Biotech GmbH Forckenbeckstraße 50 52074 Aachen Germany
- Bisy GmbH Wuenschendorf 292 Hofstätten an der Raab 8200 Hofstaetten Austria
| | - Hansjörg Weber
- Institute of Organic Chemistry NAWI Graz Graz University of Technology Stremayrgasse 9 8010 Graz Austria
| | - Wolfgang Kroutil
- Institute of Chemistry NAWI Graz University of Graz Heinrichstraße 28 8010 Graz Austria
| | - Ulrich Schwaneberg
- Institute of Biotechnology RWTH Aachen University Worringerweg 3 52074 Aachen Germany
| | - Anton Glieder
- Institute of Molecular Biotechnology NAWI Graz Graz University of Technology Petersgasse 14 8010 Graz Austria
| |
Collapse
|
26
|
In Vitro Effects of Papaverine on Cell Proliferation, Reactive Oxygen Species, and Cell Cycle Progression in Cancer Cells. Molecules 2021; 26:molecules26216388. [PMID: 34770797 PMCID: PMC8587410 DOI: 10.3390/molecules26216388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 11/23/2022] Open
Abstract
Papaverine (PPV) is an alkaloid isolated from the Papaver somniferum. Research has shown that PPV inhibits proliferation. However, several questions remain regarding the effects of PPV in tumorigenic cells. In this study, the influence of PPV was investigated on the proliferation (spectrophotometry), morphology (light microscopy), oxidative stress (fluorescent microscopy), and cell cycle progression (flow cytometry) in MDA-MB-231, A549, and DU145 cell lines. Exposure to 150 μM PPV resulted in time- and dose-dependent antiproliferative activity with reduced cell growth to 56%, 53%, and 64% in the MDA-MB-231, A549, and DU145 cell lines, respectively. Light microscopy revealed that PPV exposure increased cellular protrusions in MDA-MB-231 and A549 cells to 34% and 23%. Hydrogen peroxide production increased to 1.04-, 1.02-, and 1.44-fold in PPV-treated MDA-MB-231, A549, and DU145 cells, respectively, compared to cells propagated in growth medium. Furthermore, exposure to PPV resulted in an increase of cells in the sub-G1 phase by 46% and endoreduplication by 10% compared to cells propagated in growth medium that presented with 2.8% cells in the sub-G1 phase and less than 1% in endoreduplication. The results of this study contribute to understanding of effects of PPV on cancer cell lines.
Collapse
|
27
|
Suppression of Proliferation of Human Glioblastoma Cells by Combined Phosphodiesterase and Multidrug Resistance-Associated Protein 1 Inhibition. Int J Mol Sci 2021; 22:ijms22189665. [PMID: 34575827 PMCID: PMC8471536 DOI: 10.3390/ijms22189665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/17/2022] Open
Abstract
The paucity of currently available therapies for glioblastoma multiforme requires novel approaches to the treatment of this brain tumour. Disrupting cyclic nucleotide-signalling through phosphodiesterase (PDE) inhibition may be a promising way of suppressing glioblastoma growth. Here, we examined the effects of 28 PDE inhibitors, covering all the major PDE classes, on the proliferation of the human U87MG, A172 and T98G glioblastoma cells. The PDE10A inhibitors PF-2545920, PQ10 and papaverine, the PDE3/4 inhibitor trequinsin and the putative PDE5 inhibitor MY-5445 potently decreased glioblastoma cell proliferation. The synergistic suppression of glioblastoma cell proliferation was achieved by combining PF-2545920 and MY-5445. Furthermore, a co-incubation with drugs that block the activity of the multidrug resistance-associated protein 1 (MRP1) augmented these effects. In particular, a combination comprising the MRP1 inhibitor reversan, PF-2545920 and MY-5445, all at low micromolar concentrations, afforded nearly complete inhibition of glioblastoma cell growth. Thus, the potent suppression of glioblastoma cell viability may be achieved by combining MRP1 inhibitors with PDE inhibitors at a lower toxicity than that of the standard chemotherapeutic agents, thereby providing a new combination therapy for this challenging malignancy.
Collapse
|
28
|
Okazawa M, Oyama T, Abe H, Yamazaki H, Yoshimori A, Tsukimoto M, Yoshizawa K, Takao K, Sugita Y, Kamiya T, Uchiumi F, Sakagami H, Abe T, Tanuma SI. A 3-styrylchromone converted from trimebutine 3D pharmacophore possesses dual suppressive effects on RAGE and TLR4 signaling pathways. Biochem Biophys Res Commun 2021; 566:1-8. [PMID: 34111666 DOI: 10.1016/j.bbrc.2021.05.096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/28/2021] [Indexed: 01/22/2023]
Abstract
Receptor for advanced glycation end-products (RAGE) and Toll-like receptors (TLRs) are potential therapeutic targets in the treatment of acute and chronic inflammatory diseases. We previously reported that trimebutine, a spasmolytic drug, suppresses RAGE pro-inflammatory signaling pathway in macrophages. The aim of this study was to convert trimebutine to a new small molecule using in silico 3D pharmacophore similarity search, and dissect the mechanistic anti-inflammatory basis. Of note, a unique 3-styrylchromone (3SC), 7-methoxy-3-trimethoxy-SC (7M3TMSC), converted from trimebutine 3D pharmacophore potently suppressed both high mobility group box 1-RAGE and lipopolysaccharide-TLR4 signaling pathways in macrophage-like RAW264.7 cells. More importantly, 7M3TMSC inhibited the phosphorylation of extracellular signaling-regulated kinase 1 and 2 (ERK1/2) and downregulated the production of cytokines, such as interleukin-6. Furthermore, 3D pharmacophore-activity relationship analyses revealed that the hydrogen bond acceptors of the trimethoxy groups in a 3-styryl moiety and the 7-methoxy-group in a chromone moiety in this compound are significant in the dual anti-inflammatory activity. Thus, 7M3TMSC may provide an important scaffold for the development of a new type of anti-inflammatory dual effective drugs targeting RAGE/TLR4-ERK1/2 signaling.
Collapse
Affiliation(s)
- Miwa Okazawa
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Takahiro Oyama
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Hideaki Abe
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Hiroaki Yamazaki
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Atsushi Yoshimori
- Institute for Theoretical Medicine Inc., Fujisawa, Kanagawa, 251-0012, Japan
| | - Mitsutoshi Tsukimoto
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Koichi Takao
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Sakado, Saitama, 350-0295, Japan
| | - Yoshiaki Sugita
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Sakado, Saitama, 350-0295, Japan
| | - Takanori Kamiya
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Fumiaki Uchiumi
- Department of Gene Regulation, Fuculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Hiroshi Sakagami
- Meikai University Research Institute of Odontology (M-RIO), Sakado, Saitama, 350-0283, Japan
| | - Takehiko Abe
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Sei-Ichi Tanuma
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
29
|
An Alternative Pipeline for Glioblastoma Therapeutics: A Systematic Review of Drug Repurposing in Glioblastoma. Cancers (Basel) 2021; 13:cancers13081953. [PMID: 33919596 PMCID: PMC8073966 DOI: 10.3390/cancers13081953] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioblastoma is a devastating malignancy that has continued to prove resistant to a variety of therapeutics. No new systemic therapy has been approved for use against glioblastoma in almost two decades. This observation is particularly disturbing given the amount of money invested in identifying novel therapies for this disease. A relatively rapid and economical pipeline for identification of novel agents is drug repurposing. Here, a comprehensive review detailing the state of drug repurposing in glioblastoma is provided. We reveal details on studies that have examined agents in vitro, in animal models and in patients. While most agents have not progressed beyond the initial stages, several drugs, from a variety of classes, have demonstrated promising results in early phase clinical trials. Abstract The treatment of glioblastoma (GBM) remains a significant challenge, with outcome for most pa-tients remaining poor. Although novel therapies have been developed, several obstacles restrict the incentive of drug developers to continue these efforts including the exorbitant cost, high failure rate and relatively small patient population. Repositioning drugs that have well-characterized mechanistic and safety profiles is an attractive alternative for drug development in GBM. In ad-dition, the relative ease with which repurposed agents can be transitioned to the clinic further supports their potential for examination in patients. Here, a systematic analysis of the literature and clinical trials provides a comprehensive review of primary articles and unpublished trials that use repurposed drugs for the treatment of GBM. The findings demonstrate that numerous drug classes that have a range of initial indications have efficacy against preclinical GBM models and that certain agents have shown significant potential for clinical benefit. With examination in randomized, placebo-controlled trials and the targeting of particular GBM subgroups, it is pos-sible that repurposing can be a cost-effective approach to identify agents for use in multimodal anti-GBM strategies.
Collapse
|
30
|
A Unique Anti-Cancer 3-Styrylchromone Suppresses Inflammatory Response via HMGB1-RAGE Signaling. MEDICINES 2021; 8:medicines8040017. [PMID: 33805209 PMCID: PMC8064355 DOI: 10.3390/medicines8040017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
Background: High mobility group box 1 (HMGB1)-receptor for advanced glycation endo-products (RAGE) axis serves as a key player in linking inflammation and carcinogenesis. Recently, papaverine was revealed to suppress the HMGB1-RAGE inflammatory signaling pathway and cancer cell proliferation. Therefore, a dual suppressor targeting this axis is expected to become a new type of therapeutic agent to treat cancer. Methods: Papaverine 3D pharmacophore mimetic compounds were selected by the LigandScout software from our in-house, anti-cancer chemical library and assessed for their anti-inflammatory activities by a HMGB1-RAGE-mediated interleukin-6 production assay using macrophage-like RAW264.7 cells. Molecular-biological analyses, such as Western blotting, were performed to clarify the mechanism of action. Results: A unique 6-methoxy-3-hydroxy-styrylchromone was found to possess potent anti-inflammatory and anti-cancer activities via the suppression of the HMGB1-RAGE-extracellular signal-regulated kinase 1/2 signaling pathway. Furthermore, the 3D pharmacophore-activity relationship analyses revealed that the hydroxyl group at the C4′ position of the benzene ring in a 3-styryl moiety was significant in its dual suppressive effects. Conclusions: These findings indicated that this compound may provide a valuable scaffold for the development of a new type of anti-cancer drug possessing anti-inflammatory activity and as a tool for understanding the link between inflammation and carcinogenesis.
Collapse
|
31
|
Kurasaka C, Ogino Y, Sato A. Molecular Mechanisms and Tumor Biological Aspects of 5-Fluorouracil Resistance in HCT116 Human Colorectal Cancer Cells. Int J Mol Sci 2021; 22:ijms22062916. [PMID: 33805673 PMCID: PMC8002131 DOI: 10.3390/ijms22062916] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
5-Fluorouracil (5-FU) is a cornerstone drug used in the treatment of colorectal cancer (CRC). However, the development of resistance to 5-FU and its analogs remain an unsolved problem in CRC treatment. In this study, we investigated the molecular mechanisms and tumor biological aspects of 5-FU resistance in CRC HCT116 cells. We established an acquired 5-FU-resistant cell line, HCT116RF10. HCT116RF10 cells were cross-resistant to the 5-FU analog, fluorodeoxyuridine. In contrast, HCT116RF10 cells were collaterally sensitive to SN-38 and CDDP compared with the parental HCT16 cells. Whole-exome sequencing revealed that a cluster of genes associated with the 5-FU metabolic pathway were not significantly mutated in HCT116 or HCT116RF10 cells. Interestingly, HCT116RF10 cells were regulated by the function of thymidylate synthase (TS), a 5-FU active metabolite 5-fluorodeoxyuridine monophosphate (FdUMP) inhibiting enzyme. Half of the TS was in an active form, whereas the other half was in an inactive form. This finding indicates that 5-FU-resistant cells exhibited increased TS expression, and the TS enzyme is used to trap FdUMP, resulting in resistance to 5-FU and its analogs.
Collapse
Affiliation(s)
- Chinatsu Kurasaka
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (C.K.); (Y.O.)
| | - Yoko Ogino
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (C.K.); (Y.O.)
- Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Akira Sato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; (C.K.); (Y.O.)
- Correspondence: ; Tel.: +81-4-7121-3620
| |
Collapse
|
32
|
Beeraka NM, Bovilla VR, Doreswamy SH, Puttalingaiah S, Srinivasan A, Madhunapantula SV. The Taming of Nuclear Factor Erythroid-2-Related Factor-2 (Nrf2) Deglycation by Fructosamine-3-Kinase (FN3K)-Inhibitors-A Novel Strategy to Combat Cancers. Cancers (Basel) 2021; 13:cancers13020281. [PMID: 33466626 PMCID: PMC7828646 DOI: 10.3390/cancers13020281] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Aim of this review is to provide an overview on (a) Fructosamine-3-Kinase (FN3K) and its role in regulating Nuclear Factor Erythorid-2-Related Factor-2 (Nrf2); (b) the role of glycation and deglycation mechanisms in modulating the functional properties of proteins, in particular, the Nrf2; (c) the dual role of Nrf2 in the prevention and treatment of cancers. Since controlling the glycation of Nrf2 is one of the key mechanisms determining the fate of a cell; whether to get transformed into a cancerous one or to stay as a normal one, it is important to regulate Nrf2 and deglycating FN3K using pharmacological agents. Inhibitors of FN3K are being explored currently to modulate Nrf2 activity thereby control the cancers. Abstract Glycated stress is mediated by the advanced glycation end products (AGE) and the binding of AGEs to the receptors for advanced glycation end products (RAGEs) in cancer cells. RAGEs are involved in mediating tumorigenesis of multiple cancers through the modulation of several downstream signaling cascades. Glycated stress modulates various signaling pathways that include p38 mitogen-activated protein kinase (p38 MAPK), nuclear factor kappa–B (NF-κB), tumor necrosis factor (TNF)-α, etc., which further foster the uncontrolled proliferation, growth, metastasis, angiogenesis, drug resistance, and evasion of apoptosis in several cancers. In this review, a balanced overview on the role of glycation and deglycation in modulating several signaling cascades that are involved in the progression of cancers was discussed. Further, we have highlighted the functional role of deglycating enzyme fructosamine-3-kinase (FN3K) on Nrf2-driven cancers. The activity of FN3K is attributed to its ability to deglycate Nrf2, a master regulator of oxidative stress in cells. FN3K is a unique protein that mediates deglycation by phosphorylating basic amino acids lysine and arginine in various proteins such as Nrf2. Deglycated Nrf2 is stable and binds to small musculoaponeurotic fibrosarcoma (sMAF) proteins, thereby activating cellular antioxidant mechanisms to protect cells from oxidative stress. This cellular protection offered by Nrf2 activation, in one way, prevents the transformation of a normal cell into a cancer cell; however, in the other way, it helps a cancer cell not only to survive under hypoxic conditions but also, to stay protected from various chemo- and radio-therapeutic treatments. Therefore, the activation of Nrf2 is similar to a double-edged sword and, if not controlled properly, can lead to the development of many solid tumors. Hence, there is a need to develop novel small molecule modulators/phytochemicals that can regulate FN3K activity, thereby maintaining Nrf2 in a controlled activation state.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India; (N.M.B.); (V.R.B.); (S.H.D.); (S.P.)
| | - Venugopal R. Bovilla
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India; (N.M.B.); (V.R.B.); (S.H.D.); (S.P.)
- Public Health Research Institute of India (PHRII), Mysuru, Karnataka 570020, India
| | - Shalini H. Doreswamy
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India; (N.M.B.); (V.R.B.); (S.H.D.); (S.P.)
| | - Sujatha Puttalingaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India; (N.M.B.); (V.R.B.); (S.H.D.); (S.P.)
| | - Asha Srinivasan
- Division of Nanoscience and Technology, Faculty of Life Sciences, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India;
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India; (N.M.B.); (V.R.B.); (S.H.D.); (S.P.)
- Special Interest Group in Cancer Biology and Cancer Stem Cells, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka 570015, India
- Correspondence: ; Tel.: +91-810-527-8621
| |
Collapse
|
33
|
Gaber A, Alsanie WF, Kumar DN, Refat MS, Saied EM. Novel Papaverine Metal Complexes with Potential Anticancer Activities. Molecules 2020; 25:molecules25225447. [PMID: 33233775 PMCID: PMC7699950 DOI: 10.3390/molecules25225447] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Although several potential therapeutic agents have been developed to efficiently treat cancer, some side effects can occur simultaneously. Papaverine, a non-narcotic opium alkaloid, is a potential anticancer drug that showed selective antitumor activity in various tumor cells. Recent studies have demonstrated that metal complexes improve the biological activity of the parent bioactive ligands. Based on those facts, herein we describe the synthesis of novel papaverine–vanadium(III), ruthenium(III) and gold(III) metal complexes aiming at enhancing the biological activity of papaverine drug. The structures of the synthesized complexes were characterized by various spectroscopic methods (IR, UV–Vis, NMR, TGA, XRD, SEM). The anticancer activity of synthesized metal complexes was evaluated in vitro against two types of cancer cell lines: human breast cancer MCF-7 cells and hepatocellular carcinoma HepG-2 cells. The results revealed that papaverine-Au(III) complex, among the synthesized complexes, possess potential antimicrobial and anticancer activities. Interestingly, the anticancer activity of papaverine–Au(III) complex against the examined cancer cell lines was higher than that of the papaverine alone, which indicates that Au-metal complexation improved the anticancer activity of the parent drug. Additionally, the Au complex showed anticancer activity against the breast cancer MCF-7 cells better than that of cisplatin. The biocompatibility experiments showed that Au complex is less toxic than the papaverine drug alone with IC50 ≈ 111µg/mL. These results indicate that papaverine–Au(III) complex is a promising anticancer complex-drug which would make it a suitable candidate for further in vivo investigations.
Collapse
Affiliation(s)
- Ahmed Gaber
- Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Walaa F. Alsanie
- Department of Clinical Laboratories, College of Applied Medical Sciences, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Deo Nandan Kumar
- Department of Chemistry, Deshbandhu College, University of Delhi, Delhi 110019, India;
| | - Moamen S. Refat
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Department of Chemistry, Faculty of Science, Port Said University, Port Said 42511, Egypt
- Correspondence: (M.S.R.); (E.M.S.)
| | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
- Correspondence: (M.S.R.); (E.M.S.)
| |
Collapse
|
34
|
Yoshizawa K, Takeuchi K, Nakamura T, Ukai S, Takahashi Y, Sato A, Takasawa R, Tanuma SI. Antinociceptive activity of the novel RAGE inhibitor, papaverine, in a mouse model of chronic inflammatory pain. Synapse 2020; 75:e22188. [PMID: 32979223 DOI: 10.1002/syn.22188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/01/2020] [Accepted: 08/24/2020] [Indexed: 11/10/2022]
Abstract
Extracellular high-mobility group box 1 (HMGB1) is known to mediate the inflammatory response through pattern recognition receptors, including the receptor for advanced glycation end products (RAGE) or the toll-like receptors (TLRs). The aim of the present study was to investigate whether papaverine, a novel RAGE inhibitor, could suppress inflammatory pain in mice after several time points, which was induced by the injection of complete Freund's adjuvant (CFA). We also investigated the influence of redox modulation during a state of chronic inflammatory pain. Although papaverine did not suppress CFA-induced mechanical allodynia on Day 7, papaverine significantly suppressed CFA-induced mechanical allodynia on Days 14 and 28. In contrast, the radical scavenger N-tert-Butyl-α-phenylnitrone (PBN) suppressed mechanical allodynia in mice on Days 7 and 14, but not on Day 28. We demonstrated that the RAGE inhibitor improves mechanical allodynia in chronic inflammatory conditions. Moreover, we also found that high levels of reactive oxygen species (ROS) contributed to the early phase of CFA-induced mechanical allodynia. Precisely, lower ROS levels contributed to the inflammatory pain response via the all-thiol HMGB1/RAGE signaling pathway during the chronic state. These findings led us to propose that ROS levels modulate RAGE and/or TLR4-mediated inflammatory allodynia by regulating the concentrations of disulfide HMGB1 or all-thiol HMGB1.
Collapse
Affiliation(s)
- Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Kota Takeuchi
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Toka Nakamura
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Saki Ukai
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yukino Takahashi
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Akira Sato
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryoko Takasawa
- Laboratory of Medical Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Sei-Ichi Tanuma
- Laboratory of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Japan
| |
Collapse
|
35
|
He Z, Wang C, Xue H, Zhao R, Li G. Identification of a Metabolism-Related Risk Signature Associated With Clinical Prognosis in Glioblastoma Using Integrated Bioinformatic Analysis. Front Oncol 2020; 10:1631. [PMID: 33042807 PMCID: PMC7523182 DOI: 10.3389/fonc.2020.01631] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Altered metabolism of glucose, lipid and glutamine is a prominent hallmark of cancer cells. Currently, cell heterogeneity is believed to be the main cause of poor prognosis of glioblastoma (GBM) and is closely related to relapse caused by therapy resistance. However, the comprehensive model of genes related to glucose-, lipid- and glutamine-metabolism associated with the prognosis of GBM remains unclear, and the metabolic heterogeneity of GBM still needs to be further explored. Based on the expression profiles of 1,395 metabolism-related genes in three datasets of TCGA/CGGA/GSE, consistent cluster analysis revealed that GBM had three different metabolic status and prognostic clusters. Combining univariate Cox regression analysis and LASSO-penalized Cox regression machine learning methods, we identified a 17-metabolism-related genes risk signature associated with GBM prognosis. Kaplan-Meier analysis found that obtained signature could differentiate the prognosis of high- and low-risk patients in three datasets. Moreover, the multivariate Cox regression analysis and receiver operating characteristic curves indicated that the signature was an independent prognostic factor for GBM and had a strong predictive power. The above results were further validated in the CGGA and GSE13041 datasets, and consistent results were obtained. Gene set enrichment analysis (GSEA) suggested glycolysis gluconeogenesis and oxidative phosphorylation were significantly enriched in high- and low-risk GBM. Lastly Connectivity Map screened 54 potential compounds specific to different subgroups of GBM patients. Our study identified a novel metabolism-related gene signature, in addition the existence of three different metabolic status and two opposite biological processes in GBM were recognized, which revealed the metabolic heterogeneity of GBM. Robust metabolic subtypes and powerful risk prognostic models contributed a new perspective to the metabolic exploration of GBM.
Collapse
Affiliation(s)
- Zheng He
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Chengcheng Wang
- Department of Pharmacy, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| |
Collapse
|
36
|
Solmaz V, Kaya M, Uslu FB, Atasoy O, Erbaş O. Papaverine Has Therapeutic Potential for Sepsis-Induced Neuropathy in Rats, Possibly via the Modulation of HMGB1-RAGE Axis and Its Antioxidant Prosperities. J INVEST SURG 2020; 35:7-13. [PMID: 32842806 DOI: 10.1080/08941939.2020.1809751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 10/23/2022]
Abstract
AIM Our aim was to investigate the possible neuroprotective properties of papaverine in sepsis-induced critical illness neuropathy (SCIN) through the evaluation of various inflammatory biochemical markers, including interleukin 6 (IL-6), C-reactive protein (CRP), and tumor necrosis factor-alpha (TNF-α), and oxidative stress biomarkers, such as malondialdehyde (MDA) and lactic acid. Additionally, evaluation of the HMGB1/RAGE interactions in SCIN was another target of this research. METHOD To create a sepsis model, a procedure involving intraperitoneal injection of feces was performed on 48 rats. The rats were divided into four equal groups: sham operated, controls and those receiving 20 and 40 mg/kg/day papaverine. After five-day treatments, compound muscle action potential (CMAPs) with electroneuromyography (ENMG) was recorded in all rats. Following ENMG evaluations, the plasma levels of sRAGE, HMGB1, TNF-α, IL-6, CRP, MDA and lactic acid were measured. RESULTS TNF-α, CRP, IL-6, HMGB1, MDA, and lactic acid levels were significantly elevated in the SCIN group, and sRAGE levels were significantly decreased. In recipients of papaverine (20 and 40 mg/kg) treatment, these biochemical findings were improved. Furthermore, electrophysiological findings also showed significant improvement in both 20 and 40 mg/kg papaverine treated groups. CONCLUSION Papaverine demonstrates neuroprotective effects in a rat model of SCIN. Considering its anti-inflammatory and antioxidant properties, papaverine's neuroprotective effects possibly stem from the suppression of the RAGE-HMGB1 axis.
Collapse
Affiliation(s)
- Volkan Solmaz
- Department of Neurology, Memorial Hizmet Hospital, Istanbul, Turkey
| | - Mahmut Kaya
- Department of Internal Medicine, Memorial Hizmet Hospital, Istanbul, Turkey
| | - Fatma Betul Uslu
- Department of Anesthesiology, Batman State Hospital, Batman, Turkey
| | - Ozum Atasoy
- Radiation Oncology, Kartal Dr. Lutfi Kırdar Traning and Research Hospital, Istanbul, Turkey
| | - Oytun Erbaş
- Medical Faculty, Department of Physiology, Demiroğlu Bilim University, Istanbul, Turkey
| |
Collapse
|
37
|
Tregubenko P, Zvonarev V. Impact of Opioid Use in Hematological Malignancies: Clinical, Immunological and Concomitant Aspects. J Hematol 2020; 9:41-54. [PMID: 32855752 PMCID: PMC7430860 DOI: 10.14740/jh689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023] Open
Abstract
Opioid agents play a unique role in pain and symptom management for cancer patients. Research shows that opiate use, especially when associated with underlying cancer, has significant effects on hematological parameters. These changes may lead to greater risk for immunosuppression, tumor growth and progression of metastatic processes. The aim of this review is to explore the effects of opiates on various metabolic and biological processes, as well as the hematopoietic system, especially in cancer patients. Our findings demonstrate that the tumor-promoting effects of opiates remain contradictory, as both growth-promoting and anti-tumor effects have been observed. However, available data suggest that opiates can facilitate the proliferation and migration of tumor cells, and understanding of this process on cancer treatment is tremendously important.
Collapse
Affiliation(s)
- Polina Tregubenko
- Internal Medicine Residency Program, Albert Einstein College of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - Valeriy Zvonarev
- School of Behavioral Sciences, California Southern University, Costa Mesa, CA, USA.,Psychiatry Residency Training Program, Center for Behavioral Medicine, UMKC, 1000 E. 24th Street, Kansas City, MO 64108, USA
| |
Collapse
|
38
|
Khan MQ, Cirjan C, Quadri N, Alexopoulos G, Coppens J. Symptomatic cerebral vasospasm in the setting of carmustine wafer placement for glioblastoma: A case presentation and review of literature. Surg Neurol Int 2020; 11:168. [PMID: 32637221 PMCID: PMC7332710 DOI: 10.25259/sni_257_2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Gliadel placement in glioblastoma resection, particularly with concurrent chemoradiation, has demonstrated an improvement in survival. There have been several reported adverse effects, some of which lend to significantly increased morbidity and mortality. With only two other cases described in literature, cerebral vasospasm secondary to carmustine-impregnated wafers is an extremely rare side effect. Case Description: We report the case of a 51-year-old female who presented with the left lower limb paresis 8 days after high-grade glioma resection provoked by carmustine wafer placement. Conclusion: We urge surgeons to reconsider placement of carmustine wafers in nations where the surgical resection cavity includes exposed large cerebral vasculature. We also propose the early identification of this devastating complication in the postoperative period by maintaining a high clinical suspicion and prompt utilization of computed tomography and digital subtraction angiography in the management and treatment of these patients accordingly.
Collapse
Affiliation(s)
- Maheen Qamar Khan
- Department of Neurosurgery, Saint Louis University, 3635 Vista Avenue, St, Louis, Missouri, United States
| | - Cristian Cirjan
- Department of Neurosurgery, Saint Louis University, 3635 Vista Avenue, St, Louis, Missouri, United States
| | - Nabiha Quadri
- Department of Neurosurgery, Saint Louis University, 3635 Vista Avenue, St, Louis, Missouri, United States
| | - Georgios Alexopoulos
- Department of Neurosurgery, Saint Louis University, 3635 Vista Avenue, St, Louis, Missouri, United States
| | - Jeroen Coppens
- Department of Neurosurgery, Saint Louis University, 3635 Vista Avenue, St, Louis, Missouri, United States
| |
Collapse
|
39
|
El-Far AH, Sroga G, Al Jaouni SK, Mousa SA. Role and Mechanisms of RAGE-Ligand Complexes and RAGE-Inhibitors in Cancer Progression. Int J Mol Sci 2020; 21:ijms21103613. [PMID: 32443845 PMCID: PMC7279268 DOI: 10.3390/ijms21103613] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
Interactions of the receptor for advanced glycation end product (RAGE) and its ligands in the context of their role in diabetes mellitus, inflammation, and carcinogenesis have been extensively investigated. This review focuses on the role of RAGE-ligands and anti-RAGE drugs capable of controlling cancer progression. Different studies have demonstrated interaction of RAGE with a diverse range of acidic (negatively charged) ligands such as advanced glycation end products (AGEs), high-mobility group box1 (HMGB1), and S100s, and their importance to cancer progression. Some RAGE-ligands displayed effects on anti- and pro-apoptotic proteins through upregulation of the phosphatidylinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinases (MAPKs), matrix metalloproteinases (MMPs), vascular endothelial growth factor (VEGF), and nuclear factor kappa B (NF-κB) pathways, while downregulating p53 in cancer progression. In addition, RAGE may undergo ligand-driven multimodal dimerization or oligomerization mediated through self-association of some of its subunits. We conclude our review by proposing possible future lines of study that could result in control of cancer progression through RAGE inhibition.
Collapse
Affiliation(s)
- Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Damanhour 22511, Egypt;
| | - Grazyna Sroga
- Rensselaer Polytechnic Institute, NY (RPI), Troy, NY 12180, USA;
| | - Soad K. Al Jaouni
- Department of Hematology/Pediatric Oncology, King Abdulaziz University, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaker A. Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
- Correspondence:
| |
Collapse
|
40
|
Abstract
The rediscovery and reinterpretation of the Warburg effect in the year 2000 occulted for almost a decade the key functions exerted by mitochondria in cancer cells. Until recent times, the scientific community indeed focused on constitutive glycolysis as a hallmark of cancer cells, which it is not, largely ignoring the contribution of mitochondria to the malignancy of oxidative and glycolytic cancer cells, being Warburgian or merely adapted to hypoxia. In this review, we highlight that mitochondria are not only powerhouses in some cancer cells, but also dynamic regulators of life, death, proliferation, motion and stemness in other types of cancer cells. Similar to the cells that host them, mitochondria are capable to adapt to tumoral conditions, and probably to evolve to ‘oncogenic mitochondria' capable of transferring malignant capacities to recipient cells. In the wider quest of metabolic modulators of cancer, treatments have already been identified targeting mitochondria in cancer cells, but the field is still in infancy.
Collapse
Affiliation(s)
- Debora Grasso
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Luca X Zampieri
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Tânia Capelôa
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Justine A Van de Velde
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
41
|
Repurposing Papaverine as an Antiviral Agent against Influenza Viruses and Paramyxoviruses. J Virol 2020; 94:JVI.01888-19. [PMID: 31896588 DOI: 10.1128/jvi.01888-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Influenza viruses are highly infectious and are the leading cause of human respiratory diseases and may trigger severe epidemics and occasional pandemics. Although antiviral drugs against influenza viruses have been developed, there is an urgent need to design new strategies to develop influenza virus inhibitors due to the increasing resistance of viruses toward currently available drugs. In this study, we examined the antiviral activity of natural compounds against the following influenza virus strains: A/WSN/33 (H1N1), A/Udorn/72 (H3N2), and B/Lee/40. Papaverine (a nonnarcotic alkaloid that has been used for the treatment of heart disease, impotency, and psychosis) was found to be an effective inhibitor of multiple strains of influenza virus. Kinetic studies demonstrated that papaverine inhibited influenza virus infection at a late stage in the virus life cycle. An alteration in influenza virus morphology and viral ribonucleoprotein (vRNP) localization was observed as an effect of papaverine treatment. Papaverine is a well-known phosphodiesterase inhibitor and also modifies the mitogen-activated protein kinase (MAPK) pathway by downregulating the phosphorylation of MEK and extracellular signal-regulated kinase (ERK). Thus, the modulation of host cell signaling pathways by papaverine may be associated with the nuclear retention of vRNPs and the reduction of influenza virus titers. Interestingly, papaverine also inhibited paramyxoviruses parainfluenza virus 5 (PIV5), human parainfluenza virus 3 (HPIV3), and respiratory syncytial virus (RSV) infections. We propose that papaverine can be a potential candidate to be used as an antiviral agent against a broad range of influenza viruses and paramyxoviruses.IMPORTANCE Influenza viruses are important human pathogens that are the causative agents of epidemics and pandemics. Despite the availability of an annual vaccine, a large number of cases occur every year globally. Here, we report that papaverine, a vasodilator, shows inhibitory action against various strains of influenza virus as well as the paramyxoviruses PIV5, HPIV3, and RSV. A significant effect of papaverine on the influenza virus morphology was observed. Papaverine treatment of influenza-virus-infected cells resulted in the inhibition of virus at a later time in the virus life cycle through the suppression of nuclear export of vRNP and also interfered with the host cellular cAMP and MEK/ERK cascade pathways. This study explores the use of papaverine as an effective inhibitor of both influenza viruses as well as paramyxoviruses.
Collapse
|