1
|
Knight BJ, Grigolo TA, Tolchin ZA, Smith JM. Azine Dearomatization in Natural Product Total Synthesis. Chemistry 2025; 31:e202402413. [PMID: 39787324 DOI: 10.1002/chem.202402413] [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: 06/24/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Since antiquity, alkaloid natural products have served as medicinal ingredients that still contribute as an inspiration for the development of novel therapeutics. For the synthetic chemist, much of the importance of natural products lies in their acting as a forcing-function for the invention of new synthetic strategies and tactics for molecular assembly. With this rich history in mind, it remains an important goal for chemists to build nitrogenous structures with greater efficiency, abiding by economies of synthesis. Nitrogenous aromatic feedstocks have been an intriguing starting point for the functionalization and construction of alkaloids for several decades, but recent advances in reaction design have opened new doors for leveraging their abundance in concise synthesis. Herein, advances in this area of synthetic ingenuity will be summarized with the aim of instructing chemists towards considering dearomatization as a strategic avenue for both target-oriented and diversity-oriented synthetic campaigns. Overall, syntheses are evaluated, compared, and contrasted to give a systematic overview of this continued area of research.
Collapse
Affiliation(s)
- Brian J Knight
- Department of Medicinal Chemistry, Asha Therapeutics, 3802 Spectrum Blvd. Suite 146, Tampa, FL, 33612, USA
| | - Thiago A Grigolo
- Department of Chemistry and Biochemistry, Laboroatories of Molecular Recognition, Florida State University, 95 Chieftan Way, Tallahassee, FL, 32308, USA
| | - Zachary A Tolchin
- Department of Chemistry and Biochemistry, Laboroatories of Molecular Recognition, Florida State University, 95 Chieftan Way, Tallahassee, FL, 32308, USA
| | - Joel M Smith
- Department of Chemistry and Biochemistry, Laboroatories of Molecular Recognition, Florida State University, 95 Chieftan Way, Tallahassee, FL, 32308, USA
| |
Collapse
|
2
|
Ha B, Kang JH, Kim DH, Lee MY. Lipopolysaccharide-Induced Inflammatory Response and Its Prominent Suppression by Paspalum thunbergii Extract. Int J Mol Sci 2025; 26:1611. [PMID: 40004077 PMCID: PMC11855676 DOI: 10.3390/ijms26041611] [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: 01/14/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The extract of Paspalum thunbergii, a native perennial herb in Korea belonging to the rice family, was investigated for its anti-inflammatory activity and the underlying mechanisms driving its effects. Fifteen chemical components of the P. thunbergii extract, including rosmarinic acid and isoquercitrin, were identified using LC-MS. The extract showed antioxidative activity through DPPH and ABTS cation radical scavenging activity. The P. thunbergii extract significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in macrophage RAW 264.7 cells. The extract inhibited the expression of lipopolysaccharide-induced iNOS and COX-2, which are inflammation-related enzymes. To explore the underlying anti-inflammatory mechanism, the expression levels of signal proteins related to MAPK, NF-κB, JAK/STAT, and Wnt/β-catenin signaling were measured. As a result, the P. thunbergii extract inhibited the expression of p-p38, and p-JNK increased by LPS in RAW 264.7 cells. Additionally, it decreased the expression of LPS-induced p-IKKβ and p-NF-κB p65 and prevented the migration of p-NF-κB into the nucleus caused by LPS. Notably, p-JAK1, p-STAT3, Wnt 3α, β-catenin, and p-GSK-3β protein expressions were also inhibited. Therefore, the prominent anti-inflammatory activity of the P. thunbergii extract may be via the MAPK, NF-κB, JAK/STAT, Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Bin Ha
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Ji-Hye Kang
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Do Hyun Kim
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| |
Collapse
|
3
|
Arfeen M, Dhaked DK, Mani V. Multipotent Effect of Clozapine on Lipopolysaccharide-Induced Acetylcholinesterase, Cyclooxygenase-2,5-Lipoxygenase, and Caspase-3: In Vivo and Molecular Modeling Studies. Molecules 2025; 30:266. [PMID: 39860136 PMCID: PMC11767763 DOI: 10.3390/molecules30020266] [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: 12/05/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Dual inhibition of cyclooxygenase-2 (COX-2) and lipoxygenase (LOX) is a recognized strategy for enhanced anti-inflammatory effects in small molecules, offering potential therapeutic benefits for individuals at risk of dementia, particularly those with neurodegenerative diseases, common cancers, and diabetes type. Alzheimer's disease (AD) is the most common cause of dementia, and the inhibition of acetylcholinesterase (AChE) is a key approach in treating AD. Meanwhile, Caspase-3 catalyzes early events in apoptosis, contributing to neurodegeneration and subsequently AD. Structure-based virtual screening of US-FDA-approved molecules from the ZINC15 database identified clozapine (CLOZ) as the dual inhibitor of COX-2 and AChE, with significant binding affinity. Further molecular docking of CLOZ in the active site of LOX and Caspase-3 also showed significant binding potential. Further, the results from molecular docking were validated using molecular dynamics simulation (MDS) studies, confirming the results from molecular docking. The results from MDS showed good binding potential and interactions with key residues. The CLOZ was further assessed using lipopolysaccharide (LPS)-challenged rats treated for thirty days at doses of 5 and 10 mg/kg, p.o. The results demonstrated modulation of COX-2, 5-LOX, AChE, Caspase-3, and MDA in LPS-induced brains. Additionally, the expression level of IL-10 was also measured. Our results showed a significant decrease in the levels of COX-2, 5-LOX, AChE, Caspase-3, and MDA. Our results also showed a significant decrement in the pro-inflammatory markers NF-κB, TNF-α, and IL-6 and an improvement in the levels of anti-inflammatory markers IL-10 and TGF-β1. Overall, the findings indicate that CLOZ has potential for neuroprotective effects against LPS-treated rats and can be explored.
Collapse
Affiliation(s)
- Minhajul Arfeen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Devendra Kumar Dhaked
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata 700054, India;
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| |
Collapse
|
4
|
Zhang LL, Zhang DJ, Shi JX, Huang MY, Yu JM, Chen XJ, Wei X, Zou L, Lu JJ. Immunogenic cell death inducers for cancer therapy: An emerging focus on natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155828. [PMID: 38905847 DOI: 10.1016/j.phymed.2024.155828] [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: 04/01/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Immunogenic cell death (ICD) is a specific form of regulated cell death induced by a variety of stressors. During ICD, the dying cancer cells release damage-associated molecular patterns (DAMPs), which promote dendritic cell maturation and tumor antigen presentation, subsequently triggering a T-cell-mediated anti-tumor immune response. In recent years, a growing number of studies have demonstrated the potential of natural products to induce ICD and enhance tumor cell immunogenicity. Moreover, there is an increasing interest in identifying new ICD inducers from natural products. PURPOSE This study aimed to emphasize the potential of natural products and their derivatives as ICD inducers to promote research on using natural products in cancer therapy and provide ideas for future novel immunotherapies based on ICD induction. METHOD This review included a thorough search of the PubMed, Web of Science, Scopus, and Google Scholar databases to identify natural products with ICD-inducing capabilities. A comprehensive search for clinical trials on natural ICD inducers was also conducted using ClinicalTrials.gov, as well as the approved patents using the Espacenet and CNKI Patent Database. RESULTS Natural compounds that induce ICD can be categorized into several groups, such as polyphenols, flavonoids, terpenoids, and alkaloids. Natural products can induce the release of DAMPs by triggering endoplasmic reticulum stress, activation of autophagy-related pathways, and reactive oxygen species generation, etc. Ultimately, they activate anti-tumor immune response and improve the efficacy of cancer treatments. CONCLUSION A growing number of ICD inducers from natural products with promising anti-cancer potential have been identified. The detailed information presented in this review will contribute to the further development of natural ICD inducers and cancer treatment strategies based on ICD-induced responses.
Collapse
Affiliation(s)
- Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Du-Juan Zhang
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia-Xin Shi
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jia-Mei Yu
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xu-Jia Chen
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xiao Wei
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
5
|
Ahmad I, Khalid H, Perveen A, Shehroz M, Nishan U, Rahman FU, Sheheryar, Moura AA, Ullah R, Ali EA, Shah M, Ojha SC. Identification of Novel Quinolone and Quinazoline Alkaloids as Phosphodiesterase 10A Inhibitors for Parkinson's Disease through a Computational Approach. ACS OMEGA 2024; 9:16262-16278. [PMID: 38617664 PMCID: PMC11007772 DOI: 10.1021/acsomega.3c10351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/16/2024]
Abstract
Phosphodiesterases (PDEs) are vital in signal transduction, specifically by hydrolyzing cAMP and cGMP. Within the PDE family, PDE10A is notable for its prominence in the striatum and its regulatory function over neurotransmitters in medium-spiny neurons. Given the dopamine deficiency in Parkinson's disease (PD) that affects striatal pathways, PDE10A inhibitors could offer therapeutic benefits by modulating D1 and D2 receptor signaling. This study was motivated by the successful history of quinazoline/quinazoline scaffolds in the inhibition of PDE10A. This study involved detailed in silico evaluations through docking followed by pharmacological, pharmacophoric, and pharmacokinetic analyses, prioritizing central nervous system (CNS)-active drug criteria. Seven cyclic peptides, those featuring the quinazoline/quinazoline moiety at both termini, exhibited notably enhanced docking scores compared to those of the remaining alkaloids within the screened library. We identified 7 quinolines and 1 quinazoline including Lepadin G, Aspernigerin, CJ-13536, Aurachin A, 2-Undecyl-4(1H)-quinolone, Huajiaosimuline 3-Prenyl-4-prenyloxyquinolin-2-one, and Isaindigotone that followed the standard CNS active drug criteria. The dominant quinoline ring in our study and its related quinazoline were central to our evaluations; therefore, the pharmacophoric features of these scaffolds were highlighted. The top alkaloids met all CNS-active drug properties; while nonmutagenic and without PAINS alerts, many indicated potential hepatotoxicity. Among the compounds, Huajiaosimuline was particularly significant due to its alignment with lead-likeness and CNS-active criteria. Aspernigerin demonstrated its affinity for numerous dopamine receptors, which signifies its potential to alter dopaminergic neurotransmission that is directly related to PD. Interestingly, the majority of these alkaloids had biological targets primarily associated with G protein-coupled receptors, critical in PD pathophysiology. They exhibit superior excretion parameters and toxicity end-points compared to the standard. Notably, selected alkaloids demonstrated stability in the binding pocket of PDE10A according to the molecular dynamic simulation results. Our findings emphasize the potential of these alkaloids as PDE10A inhibitors. Further experimental studies may be necessary to confirm their actual potency in inhibiting PDE10A before exploring their therapeutic potential in PD.
Collapse
Affiliation(s)
- Iqra Ahmad
- Department
of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Hira Khalid
- Department
of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Asia Perveen
- Department
of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Muhammad Shehroz
- Department
of Bioinformatics, Kohsar University Murree, Murree 47150, Pakistan
| | - Umar Nishan
- Department
of Chemistry, Kohat University of Science
& Technology, Kohat 26000, Pakistan
| | - Faiz Ur Rahman
- Department
of Zoology, University of Shangla, Shangla 19100, Khyber Pakhtunkhwa, Pakistan
| | - Sheheryar
- Department
of Animal Science, Federal University of
Ceara, Fortaleza 60020-181, Brazil
| | - Arlindo Alencar Moura
- Department
of Animal Science, Federal University of
Ceara, Fortaleza 60020-181, Brazil
| | - Riaz Ullah
- Department
of Pharmacognosy, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Essam A. Ali
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohibullah Shah
- Department
of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Suvash Chandra Ojha
- Department
of Infectious Diseases, the Affiliated Hospital
of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
6
|
Carroll AR, Copp BR, Grkovic T, Keyzers RA, Prinsep MR. Marine natural products. Nat Prod Rep 2024; 41:162-207. [PMID: 38285012 DOI: 10.1039/d3np00061c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Covering: January to the end of December 2022This review covers the literature published in 2022 for marine natural products (MNPs), with 645 citations (633 for the period January to December 2022) referring to compounds isolated from marine microorganisms and phytoplankton, green, brown and red algae, sponges, cnidarians, bryozoans, molluscs, tunicates, echinoderms, the submerged parts of mangroves and other intertidal plants. The emphasis is on new compounds (1417 in 384 papers for 2022), together with the relevant biological activities, source organisms and country of origin. Pertinent reviews, biosynthetic studies, first syntheses, and syntheses that led to the revision of structures or stereochemistries, have been included. An analysis of NP structure class diversity in relation to biota source and biome is discussed.
Collapse
Affiliation(s)
- Anthony R Carroll
- School of Environment and Science, Griffith University, Gold Coast, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Brent R Copp
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Tanja Grkovic
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, and Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Robert A Keyzers
- Centre for Biodiscovery, and School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | | |
Collapse
|
7
|
Bishop J, Wood C, Mrowicki R, Harley J. The genome sequence of the light-bulb sea squirt, Clavelina lepadiformis (Müller, 1776). Wellcome Open Res 2023; 8:543. [PMID: 38404634 PMCID: PMC10884593 DOI: 10.12688/wellcomeopenres.20417.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 02/27/2024] Open
Abstract
We present a genome assembly from an individual Clavelina lepadiformis (the light-bulb sea squirt; Chordata; Ascidiacea; Aplousobranchia; Clavelinidae). The genome sequence is 210.1 megabases in span. Most of the assembly is scaffolded into 9 chromosomal pseudomolecules. The mitochondrial genome has also been assembled and is 14.48 kilobases in length.
Collapse
Affiliation(s)
- John Bishop
- The Marine Biological Association, Plymouth, England, UK
| | - Christine Wood
- The Marine Biological Association, Plymouth, England, UK
| | - Rob Mrowicki
- The Marine Biological Association, Plymouth, England, UK
| | - Joanna Harley
- The Marine Biological Association, Plymouth, England, UK
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Casertano M, Vito A, Aiello A, Imperatore C, Menna M. Natural Bioactive Compounds from Marine Invertebrates That Modulate Key Targets Implicated in the Onset of Type 2 Diabetes Mellitus (T2DM) and Its Complications. Pharmaceutics 2023; 15:2321. [PMID: 37765290 PMCID: PMC10538088 DOI: 10.3390/pharmaceutics15092321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is an ongoing, risky, and costly health problem that therefore always requires new treatment options. Moreover, although several drugs are available, only 36% of patients achieve glycaemic control, and patient adherence is a major obstacle. With monotherapy, T2DM and its comorbidities/complications often cannot be managed, and the concurrent administration of several hypoglycaemic drugs is required, which increases the risk of side effects. In fact, despite the efficacy of the drugs currently on the market, they generally come with serious side effects. Therefore, scientific research must always be active in the discovery of new therapeutic agents. DISCUSSION The present review highlights some of the recent discoveries regarding marine natural products that can modulate the various targets that have been identified as crucial in the establishment of T2DM disease and its complications, with a focus on the compounds isolated from marine invertebrates. The activities of these metabolites are illustrated and discussed. OBJECTIVES The paper aims to capture the relevant evidence of the great chemical diversity of marine natural products as a key tool that can advance understanding in the T2DM research field, as well as in antidiabetic drug discovery. The variety of chemical scaffolds highlighted by the natural hits provides not only a source of chemical probes for the study of specific targets involved in the onset of T2DM, but is also a helpful tool for the development of drugs that are capable of acting via novel mechanisms. Thus, it lays the foundation for the design of multiple ligands that can overcome the drawbacks of polypharmacology.
Collapse
Affiliation(s)
| | | | | | | | - Marialuisa Menna
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (A.A.); (C.I.)
| |
Collapse
|
9
|
Cooreman K, De Spiegeleer B, Van Poucke C, Vanavermaete D, Delbare D, Wynendaele E, De Witte B. Emerging pharmaceutical therapies of Ascidian-derived natural products and derivatives. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104254. [PMID: 37648122 DOI: 10.1016/j.etap.2023.104254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
In a growing multidrug-resistant environment, the identification of potential new drug candidates with an acceptable safety profile is a substantial crux in pharmaceutical discovery. This review discusses several aspects and properties of approved marine natural products derived from ascidian sources (phylum Chordata, subphylum Tunicata) and/or their deduced analogues including their biosynthetic origin, (bio)chemical preclinical assessments and known efficacy-safety profiles, clinical status in trials, but also translational developments, opportunities and final conclusions. The review also describes the preclinical assessments of a large number of other ascidian compounds that have not been involved in clinical trials yet. Finally, the emerging research on the connectivity of the ascidian hosts and their independent or obligate symbiotic guests is discussed. The review covers the latest information on the topic of ascidian-derived marine natural products over the last two decades including 2022, with the majority of publications published in the last decade.
Collapse
Affiliation(s)
- Kris Cooreman
- Aquatic Environment and Quality, Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food, Jacobsenstraat 1, BE-8400 Ostend, Belgium
| | - Bart De Spiegeleer
- Faculty of Pharmaceutical Sciences, Drug Quality and Registration Group, Ghent University, Ottergemsesteenweg 460, BE-9000 Ghent, Belgium
| | - Christof Van Poucke
- Technology and Food Science Unit, Flanders Research Institute for Agriculture, Fisheries and Food, Brusselsesteenweg 370, BE-9090 Melle, Belgium
| | - David Vanavermaete
- Aquatic Environment and Quality, Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food, Jacobsenstraat 1, BE-8400 Ostend, Belgium
| | - Daan Delbare
- Aquatic Environment and Quality, Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food, Jacobsenstraat 1, BE-8400 Ostend, Belgium
| | - Evelien Wynendaele
- Faculty of Pharmaceutical Sciences, Drug Quality and Registration Group, Ghent University, Ottergemsesteenweg 460, BE-9000 Ghent, Belgium
| | - Bavo De Witte
- Aquatic Environment and Quality, Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food, Jacobsenstraat 1, BE-8400 Ostend, Belgium.
| |
Collapse
|
10
|
Wang W, Ma F, Cheung YT, Zeng G, Zhou Y, Chen Z, Liang L, Luo T, Tong R. Marine Alkaloid Lepadins E and H Induce Ferroptosis for Cancer Chemotherapy. J Med Chem 2023; 66:11201-11215. [PMID: 37578947 DOI: 10.1021/acs.jmedchem.3c00659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Induction of ferroptosis emerges as an effective method for cancer treatment. With massive efforts to elucidate the ferroptosis mechanism, the development of new ferroptosis inducers proceeds rather slowly, with only a few small molecules identified. Herein, we report our discovery of marine alkaloid lepadins E and H as a new class of ferroptosis inducers. Our in vitro studies show that lepadins E and H exhibit significant cytotoxicity, promote p53 expression, increase ROS production and lipid peroxides, reduce SLC7A11 and GPX4 levels, and upregulate ACSL4 expression, all of which consistently support induction of ferroptosis through the classical p53-SLC7A11-GPX4 pathway. Our animal model study of lepadin H confirms its in vivo antitumor efficacy with negligible toxicity to normal organs. This work elucidates the mode of action of lepadins (E and H) and verifies their in vivo efficacy as a new class of ferroptosis inducers for anticancer therapy with translational potential.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
| | - Foqing Ma
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Yuen Tsz Cheung
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Guihua Zeng
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
| | - Yiqin Zhou
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Zijing Chen
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Ministry of Education and Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Lixin Liang
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Tuoping Luo
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Ministry of Education and Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Rongbiao Tong
- Department of Chemistry, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| |
Collapse
|
11
|
Casertano M, Genovese M, Santi A, Pranzini E, Balestri F, Piazza L, Del Corso A, Avunduk S, Imperatore C, Menna M, Paoli P. Evidence of Insulin-Sensitizing and Mimetic Activity of the Sesquiterpene Quinone Avarone, a Protein Tyrosine Phosphatase 1B and Aldose Reductase Dual Targeting Agent from the Marine Sponge Dysidea avara. Pharmaceutics 2023; 15:pharmaceutics15020528. [PMID: 36839851 PMCID: PMC9964544 DOI: 10.3390/pharmaceutics15020528] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/18/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disease characterized by impaired glucose homeostasis and serious long-term complications. First-line therapeutic options for T2DM treatment are monodrug therapies, often replaced by multidrug therapies to ensure that non-responding patients maintain target glycemia levels. The use of multitarget drugs instead of mono- or multidrug therapies has been emerging as a main strategy to treat multifactorial diseases, including T2DM. Therefore, modern drug discovery in its early stages aims to identify potential modulators for multiple targets; for this purpose, exploration of the chemical space of natural products represents a powerful tool. Our study demonstrates that avarone, a sesquiterpene quinone obtained from the sponge Dysidea avara, is capable of inhibiting in vitro PTP1B, the main negative regulator of the insulin receptor, while it improves insulin sensitivity, and mitochondria activity in C2C12 cells. We observe that when avarone is administered alone, it acts as an insulin-mimetic agent. In addition, we show that avarone acts as a tight binding inhibitor of aldose reductase (AKR1B1), the enzyme involved in the development of diabetic complications. Overall, avarone could be proposed as a novel natural hit to be developed as a multitarget drug for diabetes and its pathological complications.
Collapse
Affiliation(s)
- Marcello Casertano
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
- Interdepartmental Research Center for Marine Pharmacology, Via Bonanno 6, 56126 Pisa, Italy
| | - Lucia Piazza
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
- Interdepartmental Research Center for Marine Pharmacology, Via Bonanno 6, 56126 Pisa, Italy
| | - Sibel Avunduk
- Medical Laboratory Programme, Vocational School of Health Care, Mugla University, Marmaris 48187, Turkey
| | - Concetta Imperatore
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Marialuisa Menna
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (M.M.); (P.P.); Tel.: +39-081678518 (M.M.); +39-0552751248 (P.P.)
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Correspondence: (M.M.); (P.P.); Tel.: +39-081678518 (M.M.); +39-0552751248 (P.P.)
| |
Collapse
|