1
|
Mokuolu OA, Ambrose GO, Mohammed Baba Abdulkadir, Ibrahim S, Funsho II, Mokuolu T. Exploring the genetic progression of MDR1 in Plasmodium falciparum: A decade of multi-regional genetic analysis (2014-2024). CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100304. [PMID: 39534723 PMCID: PMC11554628 DOI: 10.1016/j.crmicr.2024.100304] [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] [Indexed: 11/16/2024] Open
Abstract
Background The genetic progression of the MDR1 gene in Plasmodium falciparum, a key factor in drug resistance, presents significant challenges for malaria control. This study aims to elucidate the genetic diversity and evolutionary dynamics of P. falciparum, particularly focusing on the MDR1 gene across multi-regional populations. To analyze the genetic diversity of P. falciparum MDR1 gene across various multi-regional populations between 2014 and 2024, assessing allelic richness, genetic distances, and evolutionary patterns. Methods We conducted an extensive genetic analysis using methods such as Analysis of Molecular Variance (AMOVA), pairwise population matrices of Nei unbiased genetic distance and identity, PhiPT and Phi'PT values, and Principal Coordinates Analysis (PCoA). The study covered diverse P. falciparum populations from India, Nigeria, Ethiopia, Honduras, China, and Cameroon. Findings Our findings reveal significant genetic heterogeneity in the MDR1 gene. Populations like India: Odisha (2014) exhibited high allelic richness, indicating diverse drug resistance profiles. Notable genetic divergence was observed, especially between India (2016) and Nigeria (2020), suggesting different evolutionary trajectories in drug resistance. The PCoA analysis highlighted the multi-dimensional genetic variation, reflecting the complex interplay of factors influencing drug resistance in P. falciparum. Interpretation The comprehensive analysis of P. falciparum's MDR1 gene provides crucial insights into the multi-regional patterns of drug resistance. This knowledge is essential for developing effective malaria control measures and adapting treatment strategies to the evolving genetic diversity of the parasite.
Collapse
Affiliation(s)
- Olugbenga Ayodeji Mokuolu
- Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - George Oche Ambrose
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Mohammed Baba Abdulkadir
- Department of Paediatrics and Child Health, University of Ilorin, Ilorin, Nigeria
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Selimat Ibrahim
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Itiolu Ibilola Funsho
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Toluwani Mokuolu
- Centre for Malaria and Other Tropical Diseases Care, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| |
Collapse
|
2
|
Andrés-Rodríguez J, González-Montero MC, García-Fernández N, Calvo-Álvarez E, Pérez-Pertejo MY, Reguera-Torres RM, Balaña-Fouce R, García-Estrada C. Free Radical Production Induced by Nitroimidazole Compounds Lead to Cell Death in Leishmania infantum Amastigotes. Molecules 2024; 29:4041. [PMID: 39274889 PMCID: PMC11396368 DOI: 10.3390/molecules29174041] [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: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Leishmania infantum is the vector-borne trypanosomatid parasite causing visceral leishmaniasis in the Mediterranean basin. This neglected tropical disease is treated with a limited number of obsolete drugs that are not exempt from adverse effects and whose overuse has promoted the emergence of resistant pathogens. In the search for novel antitrypanosomatid molecules that help overcome these drawbacks, drug repurposing has emerged as a good strategy. Nitroaromatic compounds have been found in drug discovery campaigns as promising antileishmanial molecules. Fexinidazole (recently introduced for the treatment of stages 1 and 2 of African trypanosomiasis), and pretomanid, which share the nitroimidazole nitroaromatic structure, have provided antileishmanial activity in different studies. In this work, we have tested the in vitro efficacy of these two nitroimidazoles to validate our 384-well high-throughput screening (HTS) platform consisting of L. infantum parasites emitting the near-infrared fluorescent protein (iRFP) as a biomarker of cell viability. These molecules showed good efficacy in both axenic and intramacrophage amastigotes and were poorly cytotoxic in RAW 264.7 and HepG2 cultures. Fexinidazole and pretomanid induced the production of ROS in axenic amastigotes but were not able to inhibit trypanothione reductase (TryR), thus suggesting that these compounds may target thiol metabolism through a different mechanism of action.
Collapse
Affiliation(s)
- Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Estefanía Calvo-Álvarez
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - María-Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa-María Reguera-Torres
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
3
|
Mayer AMS, Mayer VA, Swanson-Mungerson M, Pierce ML, Rodríguez AD, Nakamura F, Taglialatela-Scafati O. Marine Pharmacology in 2019-2021: Marine Compounds with Antibacterial, Antidiabetic, Antifungal, Anti-Inflammatory, Antiprotozoal, Antituberculosis and Antiviral Activities; Affecting the Immune and Nervous Systems, and Other Miscellaneous Mechanisms of Action. Mar Drugs 2024; 22:309. [PMID: 39057418 PMCID: PMC11278370 DOI: 10.3390/md22070309] [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: 05/22/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The current 2019-2021 marine pharmacology literature review provides a continuation of previous reviews covering the period 1998 to 2018. Preclinical marine pharmacology research during 2019-2021 was published by researchers in 42 countries and contributed novel mechanism-of-action pharmacology for 171 structurally characterized marine compounds. The peer-reviewed marine natural product pharmacology literature reported antibacterial, antifungal, antiprotozoal, antituberculosis, and antiviral mechanism-of-action studies for 49 compounds, 87 compounds with antidiabetic and anti-inflammatory activities that also affected the immune and nervous system, while another group of 51 compounds demonstrated novel miscellaneous mechanisms of action, which upon further investigation, may contribute to several pharmacological classes. Thus, in 2019-2021, a very active preclinical marine natural product pharmacology pipeline provided novel mechanisms of action as well as new lead chemistry for the clinical marine pharmaceutical pipeline targeting the therapy of several disease categories.
Collapse
Affiliation(s)
- Alejandro M. S. Mayer
- Department of Pharmacology, College of Graduate Studies, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA;
| | - Veronica A. Mayer
- Department of Nursing Education, School of Nursing, Aurora University, 347 S. Gladstone Ave., Aurora, IL 60506, USA;
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA;
| | - Marsha L. Pierce
- Department of Pharmacology, College of Graduate Studies, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA;
| | - Abimael D. Rodríguez
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan, PR 00926, USA;
| | - Fumiaki Nakamura
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku 169-8555, Tokyo, Japan;
| | | |
Collapse
|
4
|
Amador LA, Colón-Lorenzo EE, Rodríguez AD, Serrano AE. Probing the Antiplasmodial Properties of Plakortinic Acids C and D: An Uncommon Pair of Marine Peroxide-Polyketides Isolated from a Two-Sponge Association of Plakortis symbiotica and Xetospongia deweerdtae Collected near Puerto Rico. Life (Basel) 2024; 14:684. [PMID: 38929667 PMCID: PMC11204963 DOI: 10.3390/life14060684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Plakortinic acids C (1) and D (2), an unseparable pair of endoperoxide polyketides isolated and purified from the symbiotic association of Caribbean Sea sponges Plakortis symbiotica-Xestospongia deweerdtae, underwent in vitro evaluation for antiplasmodial activity against the malaria parasite Plasmodium berghei using a drug luminescence assay. Initial screening at 10 µM revealed 50% in vitro parasite growth inhibition. The title compounds displayed antiplasmodial activity with an EC50 of 5.3 µM toward P. berghei parasites. The lytic activity against erythrocytes was assessed through an erythrocyte cell lysis assay, which showed non-lytic activity at lower concentrations ranging from 1.95 to 3.91 µM. The antiplasmodial activity and the absence of hemolytic activity support the potential of plakortinic acids C (1) and D (2) as promising lead compounds. Moreover, drug-likeness (ADMET) properties assessed through the pkCSM server predicted high intestinal absorption, hepatic metabolism, and volume of distribution, indicating favorable pharmacokinetic profiles for oral administration. These findings suggest the potential suitability of these metabolites for further investigations of antiplasmodial activity in multiple parasitic stages in the mosquito and Plasmodium falciparum. Notably, this study represents the first report of a marine natural product exhibiting the unique 7,8-dioxatricyclo[4.2.2.02,5]dec-9-ene motif being evaluated against malaria.
Collapse
Affiliation(s)
- Luis A. Amador
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan 00926, Puerto Rico;
| | - Emilee E. Colón-Lorenzo
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan 00921, Puerto Rico;
| | - Abimael D. Rodríguez
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce de León Avenue, San Juan 00926, Puerto Rico;
| | - Adelfa E. Serrano
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan 00921, Puerto Rico;
| |
Collapse
|
5
|
Utami PD, Setianingsih H, Sari DRT. In Silico Approach Triterpene Glycoside of H. atra Targeting Orotidine 5-Monophosphate Decarboxylase Protein (PfOMPDC) in P. falciparum Infection Mechanism. BIOMED RESEARCH INTERNATIONAL 2024; 2024:5924799. [PMID: 38590385 PMCID: PMC11001475 DOI: 10.1155/2024/5924799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/31/2023] [Accepted: 02/12/2024] [Indexed: 04/10/2024]
Abstract
This study accessed the potential antimalarial activity of triterpene glycoside of H. atra through targeting orotidine 5-monophosphate decarboxylase protein (PfOMPDC) in P. falciparum by molecular docking. Nine triterpene glycosides from H. atra extract modeled the structure by the Corina web server and interacted with PfOMPDC protein by using Hex 8.0.0. The docking results were visualized and analyzed by Discovery Studio version 21.1.1. 17-Hydroxyfuscocineroside B showed the lowest binding energy in PfOMPDC interaction, which was -1,098.13 kJ/mol. Holothurin A3, echinoside A, and fuscocineroside C showed low binding energy. Nine triterpene glycosides of H. atra performed interaction with PfOMPDC protein at the same region. Holothurin A1 posed interaction with PfOMPDC protein by 8 hydrogen bonds, 3 hydrophobic interactions, and 8 unfavorable bonds. Several residues were detected in the same active sites of other triterpene glycosides. Residue TYR111 was identified in all triterpene glycoside complexes, except holothurin A3 and calcigeroside B. In summary, the triterpene glycoside of H. atra is potentially a drug candidate for malaria therapeutic agents. In vitro and in vivo studies were required for further investigation.
Collapse
Affiliation(s)
- Prawesty Diah Utami
- Parasitology Departement, Faculty of Medicine, Hang Tuah University, Surabaya, Indonesia
| | - Herin Setianingsih
- Anatomy and Histology Departement, Faculty of Medicine, Hang Tuah University, Surabaya, Indonesia
| | - Dewi Ratih Tirto Sari
- Pharmacy Department, Faculty of Medical Science, Ibrahimy University, Situbondo, Indonesia
| |
Collapse
|
6
|
Cheng W, Huang Y, Gao H, Bold B, Zhang T, Yang D. Marine Natural Products as Novel Treatments for Parasitic Diseases. Handb Exp Pharmacol 2024. [PMID: 38554166 DOI: 10.1007/164_2024_712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
Parasitic diseases including malaria, leishmaniasis, and trypanosomiasis have received significant attention due to their severe health implications, especially in developing countries. Marine natural products from a vast and diverse range of marine organisms such as sponges, corals, molluscs, and algae have been found to produce unique bioactive compounds that exhibit promising potent properties, including antiparasitic, anti-Plasmodial, anti-Leishmanial, and anti-Trypanosomal activities, providing hope for the development of effective treatments. Furthermore, various techniques and methodologies have been used to investigate the mechanisms of these antiparasitic compounds. Continued efforts in the discovery and development of marine natural products hold significant promise for the future of novel treatments against parasitic diseases.
Collapse
Affiliation(s)
- Wenbing Cheng
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, Guangxi, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yanbing Huang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, Guangxi, China
| | - Haijun Gao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
- Chengdu Fifth People's Hospital (Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine/The Second Clinical Medical College), Chengdu, Sichuan, China
| | - Bolor Bold
- National Center for Zoonotic Disease, Ulaanbaatar, Mongolia
| | - Ting Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China.
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China.
| | - Dengfeng Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, Guangxi, China
- College of Food and Quality Engineering, Nanning University, Nanning, China
| |
Collapse
|
7
|
Fermiano MH, das Neves AR, da Silva F, Barros MSA, Vieira CB, Stein AL, Frizon TEA, Braga AL, de Arruda CCP, Parisotto EB, Saba S, Rafique J, Riul TB. Selenium-Containing (Hetero)Aryl Hybrids as Potential Antileishmanial Drug Candidates: In Vitro Screening against L. amazonensis. Biomedicines 2024; 12:213. [PMID: 38255318 PMCID: PMC10812941 DOI: 10.3390/biomedicines12010213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Leishmaniasis remains a significant global health concern, with current treatments relying on outdated drugs associated with high toxicity, lengthy administration, elevated costs, and drug resistance. Consequently, the urgent need for safer and more effective therapeutic options in leishmaniasis treatment persists. Previous research has highlighted selenium compounds as promising candidates for innovative leishmaniasis therapy. In light of this, a library of 10 selenium-containing diverse compounds was designed and evaluated in this study. These compounds included selenium-substituted indole, coumarin, chromone, oxadiazole, imidazo[1,2-a]pyridine, Imidazo[2,1-b]thiazole, and oxazole, among others. These compounds were screened against Leishmania amazonensis promastigotes and intracellular amastigotes, and their cytotoxicity was assessed in peritoneal macrophages, NIH/3T3, and J774A.1 cells. Among the tested compounds, MRK-106 and MRK-108 displayed the highest potency against L. amazonensis promastigotes with reduced cytotoxicity. Notably, MRK-106 and MRK-108 exhibited IC50 values of 3.97 µM and 4.23 µM, respectively, and most of the tested compounds showed low cytotoxicity in host cells (CC50 > 200 µM). Also, compounds MRK-107 and MRK-113 showed activity against intracellular amastigotes (IC50 18.31 and 15.93 µM and SI 12.55 and 10.92, respectively). In conclusion, the identified selenium-containing compounds hold potential structures as antileishmanial drug candidates to be further explored in subsequent studies. These findings represent a significant step toward the development of safer and more effective therapies for leishmaniasis, addressing the pressing need for novel and improved treatments.
Collapse
Affiliation(s)
- Maria Helena Fermiano
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Amarith Rodrigues das Neves
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Fernanda da Silva
- Instituto de Biociências (INBIO), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil
| | | | - Camila Barbosa Vieira
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - André L. Stein
- Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Cuiabá 78060-900, MT, Brazil
| | - Tiago Elias Allievi Frizon
- Departamento de Energia e Sustentabilidade, Universidade Federal de Santa Catarina (UFSC), Campus Araranguá, Araranguá 88905-120, SC, Brazil
| | - Antonio Luiz Braga
- Departamento de Química, Universidade Federal de Santa Catarina (UFSC), Florianópolis 88040-970, SC, Brazil
| | - Carla Cardozo Pinto de Arruda
- Instituto de Biociências (INBIO), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil
| | - Eduardo Benedetti Parisotto
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| | - Sumbal Saba
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - Jamal Rafique
- Instituto de Química (INQUI), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79074-460, MS, Brazil;
- LABSO, Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil (S.S.)
| | - Thalita Bachelli Riul
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil (A.R.d.N.)
| |
Collapse
|
8
|
Barzkar N, Sukhikh S, Babich O. Study of marine microorganism metabolites: new resources for bioactive natural products. Front Microbiol 2024; 14:1285902. [PMID: 38260902 PMCID: PMC10800913 DOI: 10.3389/fmicb.2023.1285902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024] Open
Abstract
The marine environment has remained a source of novel biological molecules with diversified applications. The ecological and biological diversity, along with a unique physical environment, have provided the evolutionary advantage to the plant, animals and microbial species thriving in the marine ecosystem. In light of the fact that marine microorganisms frequently interact symbiotically or mutualistically with higher species including corals, fish, sponges, and algae, this paper intends to examine the potential of marine microorganisms as a niche for marine bacteria. This review aims to analyze and summarize modern literature data on the biotechnological potential of marine fungi and bacteria as producers of a wide range of practically valuable products (surfactants, glyco-and lipopeptides, exopolysaccharides, enzymes, and metabolites with different biological activities: antimicrobial, antitumor, and cytotoxic). Hence, the study on bioactive secondary metabolites from marine microorganisms is the need of the hour. The scientific novelty of the study lies in the fact that for the first time, the data on new resources for obtaining biologically active natural products - metabolites of marine bacteria and fungi - were generalized. The review investigates the various kinds of natural products derived from marine microorganisms, specifically focusing on marine bacteria and fungi as a valuable source for new natural products. It provides a summary of the data regarding the antibacterial, antimalarial, anticarcinogenic, antibiofilm, and anti-inflammatory effects demonstrated by marine microorganisms. There is currently a great need for scientific and applied research on bioactive secondary metabolites of marine microorganisms from the standpoint of human and animal health.
Collapse
Affiliation(s)
- Noora Barzkar
- Department of Agro-Industrial Technology, Faculty of Applied Science, Food and Agro-Industrial Research Center, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Stanislav Sukhikh
- Research and Education Center “Industrial Biotechnologies”, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Olga Babich
- Research and Education Center “Industrial Biotechnologies”, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| |
Collapse
|
9
|
Porta EOJ, Kalesh K, Steel PG. Navigating drug repurposing for Chagas disease: advances, challenges, and opportunities. Front Pharmacol 2023; 14:1233253. [PMID: 37576826 PMCID: PMC10416112 DOI: 10.3389/fphar.2023.1233253] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Chagas disease is a vector-borne illness caused by the protozoan parasite Trypanosoma cruzi (T. cruzi). It poses a significant public health burden, particularly in the poorest regions of Latin America. Currently, there is no available vaccine, and chemotherapy has been the traditional treatment for Chagas disease. However, the treatment options are limited to just two outdated medicines, nifurtimox and benznidazole, which have serious side effects and low efficacy, especially during the chronic phase of the disease. Collectively, this has led the World Health Organization to classify it as a neglected disease. To address this problem, new drug regimens are urgently needed. Drug repurposing, which involves the use of existing drugs already approved for the treatment of other diseases, represents an increasingly important option. This approach offers potential cost reduction in new drug discovery processes and can address pharmaceutical bottlenecks in the development of drugs for Chagas disease. In this review, we discuss the state-of-the-art of drug repurposing approaches, including combination therapy with existing drugs, to overcome the formidable challenges associated with treating Chagas disease. Organized by original therapeutic area, we describe significant recent advances, as well as the challenges in this field. In particular, we identify candidates that exhibit potential for heightened efficacy and reduced toxicity profiles with the ultimate objective of accelerating the development of new, safe, and effective treatments for Chagas disease.
Collapse
Affiliation(s)
| | - Karunakaran Kalesh
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Darlington, United Kingdom
| | - Patrick G. Steel
- Department of Chemistry, Durham University, Durham, United Kingdom
| |
Collapse
|
10
|
Elamin EM, Eshage SE, Mohmmode SM, Mukhtar RM, Mahjoub M, Sadelin E, Shoaib TH, Edris A, Elshamly EM, Makki AA, Ashour A, Sherif AE, Osman W, Ibrahim SRM, Mohamed GA, Alzain AA. Discovery of dual-target natural antimalarial agents against DHODH and PMT of Plasmodium falciparum: pharmacophore modelling, molecular docking, quantum mechanics, and molecular dynamics simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:709-728. [PMID: 37665563 DOI: 10.1080/1062936x.2023.2251876] [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: 07/02/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Malaria is a lethal disease that claims thousands of lives worldwide annually. The objective of this study was to identify new natural compounds that can target two P. falciparum enzymes; P. falciparum Dihydroorotate dehydrogenase (PfDHODH) and P. falciparum phosphoethanolamine methyltransferase (PfPMT). To accomplish this, e-pharmacophore modelling and molecular docking were employed against PfDHODH. Following this, 1201 natural compounds with docking scores of ≤ -7 kcal/mol were docked into the active site of the second enzyme PMT. The top nine compounds were subjected to further investigation using MM-GBSA free binding energy calculations and ADME analysis. The results revealed favourable free binding energy values better than the references, as well as acceptable pharmacokinetic properties. Compounds ZINC000013377887, ZINC000015113777, and ZINC000085595753 were scrutinized to assess their interaction stability with the PfDHODH enzyme, and chemical stability reactivity using molecular dynamics (MD) simulation and density functional theory (DFT) calculations. These findings indicate that the three natural compounds are potential candidates for dual PfDHODH and PfPMT inhibitors for malaria treatment.
Collapse
Affiliation(s)
- E M Elamin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S E Eshage
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S M Mohmmode
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - R M Mukhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - M Mahjoub
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - E Sadelin
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - T H Shoaib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - A Edris
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - E M Elshamly
- Department of Molecular Biotechnology, Hochschule Anhalt, Köthen, Germany
| | - A A Makki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - A Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Al Mansurah, Egypt
| | - A E Sherif
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Al Mansurah, Egypt
| | - W Osman
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - S R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - G A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| |
Collapse
|
11
|
Afonso RC, Yien RMK, de Siqueira LBDO, Simas NK, Dos Santos Matos AP, Ricci-Júnior E. Promising natural products for the treatment of cutaneous leishmaniasis: A review of in vitro and in vivo studies. Exp Parasitol 2023; 251:108554. [PMID: 37268108 DOI: 10.1016/j.exppara.2023.108554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/04/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Although there are available treatments for cutaneous leishmaniasis (CL), the drugs used are far from ideal, toxic, and costly, in addition to the challenge faced by the development of resistance. Plants have been used as a source of natural compounds with antileishmanial action. However, few have reached the market and become phytomedicines with registration in regulatory agencies. Difficulties related to the extraction, purification, chemical identification, efficacy, safety, and production in sufficient quantity for clinical studies, hinder the emergence of new effective phytomedicines against leishmaniasis. Despite the difficulties reported, the major research centers in the world see that natural products are a trend concerning the treatment of leishmaniasis. The present work consists of a literature review of articles with in vivo studies, covering the period from January 2011 to December 2022, providing an overview of promising natural products for CL treatment. The papers show encouraging antileishmanial action of natural compounds with reduced parasite load and lesion size in animal models, suggesting new strategies for the treatment of the disease. The results reported in this review show advances in using natural products as safe and effective formulations, which can stimulate clinical studies to establish clinical therapy. In conclusion, the information in this review article serves as a preliminary basis for establishing a therapeutic protocol for future clinical trials that can validate the safety and efficacy of natural compounds, providing the development of affordable and safe phytomedicines for the treatment of CL.
Collapse
Affiliation(s)
- Rhuane Coutinho Afonso
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil
| | - Raíssa Mara Kao Yien
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil; Laboratory of Natural Products and Biological Assays, Department of Natural Products and Food, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Naomi Kato Simas
- Laboratory of Natural Products and Biological Assays, Department of Natural Products and Food, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Paula Dos Santos Matos
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil
| | - Eduardo Ricci-Júnior
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
12
|
García-Davis S, López-Arencibia A, Bethencourt-Estrella CJ, San Nicolás-Hernández D, Viveros-Valdez E, Díaz-Marrero AR, Fernández JJ, Lorenzo-Morales J, Piñero JE. Laurequinone, a Lead Compound against Leishmania. Mar Drugs 2023; 21:333. [PMID: 37367658 DOI: 10.3390/md21060333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/28/2023] Open
Abstract
Among neglected tropical diseases, leishmaniasis is one of the leading causes, not only of deaths but also of disability-adjusted life years. This disease, caused by protozoan parasites of the genus Leishmania, triggers different clinical manifestations, with cutaneous, mucocutaneous, and visceral forms. As existing treatments for this parasitosis are not sufficiently effective or safe for the patient, in this work, different sesquiterpenes isolated from the red alga Laurencia johnstonii have been studied for this purpose. The different compounds were tested in vitro against the promastigote and amastigote forms of Leishmania amazonensis. Different assays were also performed, including the measurement of mitochondrial potential, determination of ROS accumulation, and chromatin condensation, among others, focused on the detection of the cell death process known in this type of organism as apoptosis-like. Five compounds were identified that displayed leishmanicidal activity: laurequinone, laurinterol, debromolaurinterol, isolaurinterol, and aplysin, showing IC50 values against promastigotes of 1.87, 34.45, 12.48, 10.09, and 54.13 µM, respectively. Laurequinone was the most potent compound tested and was shown to be more effective than the reference drug miltefosine against promastigotes. Different death mechanism studies carried out showed that laurequinone appears to induce programmed cell death or apoptosis in the parasite studied. The obtained results underline the potential of this sesquiterpene as a novel anti-kinetoplastid therapeutic agent.
Collapse
Affiliation(s)
- Sara García-Davis
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Spain
- Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez S/N, 38206 La Laguna, Tenerife, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), |Instituto de Salud Carlos III, 28006 Madrid, Madrid, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Ezequiel Viveros-Valdez
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Avenida Pedro de Alba S/N, San Nicolás de los Garza 66450, Nuevo León, Mexico
| | - Ana R Díaz-Marrero
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Spain
- Instituto de Productos Naturales y Agrobiología (IPNA), Consejo Superior de Investigaciones Científicas (CSIC), Avenida Astrofísico Francisco Sánchez 3, 38206 La Laguna, Tenerife, Spain
| | - José J Fernández
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Spain
- Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez S/N, 38206 La Laguna, Tenerife, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), |Instituto de Salud Carlos III, 28006 Madrid, Madrid, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez S/N, 38206 La Laguna, Tenerife, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), |Instituto de Salud Carlos III, 28006 Madrid, Madrid, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| |
Collapse
|
13
|
Keller JG, Petersen KV, Mizielinski K, Thiesen C, Bjergbæk L, Reguera RM, Pérez-Pertejo Y, Balaña-Fouce R, Trejo A, Masdeu C, Alonso C, Knudsen BR, Tesauro C. Gel-Free Tools for Quick and Simple Screening of Anti-Topoisomerase 1 Compounds. Pharmaceuticals (Basel) 2023; 16:ph16050657. [PMID: 37242440 DOI: 10.3390/ph16050657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
With the increasing need for effective compounds against cancer or pathogen-borne diseases, the development of new tools to investigate the enzymatic activity of biomarkers is necessary. Among these biomarkers are DNA topoisomerases, which are key enzymes that modify DNA and regulate DNA topology during cellular processes. Over the years, libraries of natural and synthetic small-molecule compounds have been extensively investigated as potential anti-cancer, anti-bacterial, or anti-parasitic drugs targeting topoisomerases. However, the current tools for measuring the potential inhibition of topoisomerase activity are time consuming and not easily adaptable outside specialized laboratories. Here, we present rolling circle amplification-based methods that provide fast and easy readouts for screening of compounds against type 1 topoisomerases. Specific assays for the investigation of the potential inhibition of eukaryotic, viral, or bacterial type 1 topoisomerase activity were developed, using human topoisomerase 1, Leishmania donovani topoisomerase 1, monkeypox virus topoisomerase 1, and Mycobacterium smegmatis topoisomerase 1 as model enzymes. The presented tools proved to be sensitive and directly quantitative, paving the way for new diagnostic and drug screening protocols in research and clinical settings.
Collapse
Affiliation(s)
| | | | | | - Celine Thiesen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lotte Bjergbæk
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Rosa M Reguera
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, University of León, 24071 León, Spain
| | - Angela Trejo
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Carme Masdeu
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Concepcion Alonso
- Department of Organic Chemistry, Faculty of Pharmacy, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Birgitta R Knudsen
- VPCIR Biosciences ApS, 8000 Aarhus C, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | | |
Collapse
|
14
|
de Araújo JIF, Aires NL, Almeida-Neto FWQ, Marinho MM, Marinho EM, Paula Magalhães E, de Menezes RRPPB, Sampaio TL, Maria Costa Martins A, Teixeira EH, Rafaela Freitas Dotto A, Amaral WD, Teixeira AMR, de Lima-Neto P, Marinho ES, Dos Santos HS. Antiproliferative activity on Trypanosoma cruzi (Y strain) of the triterpene 3β,6β,16β-trihidroxilup-20 (29)-ene isolated from Combretum leprosum. J Biomol Struct Dyn 2022; 40:12302-12315. [PMID: 34436980 DOI: 10.1080/07391102.2021.1970025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chagas disease infects approximately seven million people worldwide. Benznidazole is effective only in the acute phase of the disease, with an average cure rate of 80% between acute and recent cases. Therefore, there is an urgent need to find new bioactive substances that can be effective against parasites without causing so many complications to the host. In this study, the triterpene 3β-6β-16β-trihydroxilup-20 (29)-ene (CLF-1) was isolated from Combretum leprosum, and its molecular structure was determined by NMR and infrared spectroscopy. The CLF-1 was also evaluated in vitro and in silico as potential trypanocidal agent against epimastigote and trypomastigote forms of Trypanosoma cruzi (Y strain). The CLF-1 demonstrated good results highlighted by lower IC50 (76.0 ± 8.72 µM, 75.1 ± 11.0 µM, and 70.3 ± 45.4 µM) for epimastigotes at 24, 48 and 72 h, and LC50 (71.6 ± 11.6 µM) for trypomastigotes forms. The molecular docking study shows that the CLF-1 was able to interact with important TcGAPDH residues, suggesting that this natural compound may preferentially exert its effect by compromising the glycolytic pathway in T. cruzi. The ADMET study together with the MTT results indicated that the CLF-1 is well-absorbed in the intestine and has low toxicity. Thus, this work adds new evidence that CLF-1 can potentially be used as a candidate for the development of new options for the treatment of Chagas disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- José Ismael F de Araújo
- Programa de Pós-Graduação em Biotecnologia - PPGB-Renorbio, Universidade Estadual do Ceará, Fortaleza, CE, Brazil
| | - Natália L Aires
- Laboratório de Bioprospecção Farmacêutica e Bioquímica Clínica (LBFBC), Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, Brazil
| | | | - Márcia M Marinho
- Faculdade de Educação, Ciência e Letras de Iguatu, Universidade Estadual do Ceará, Iguatu, Brazil
| | - Emanuelle M Marinho
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Emanuel Paula Magalhães
- Laboratório de Bioprospecção Farmacêutica e Bioquímica Clínica (LBFBC), Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Ramon R P P B de Menezes
- Laboratório de Bioprospecção Farmacêutica e Bioquímica Clínica (LBFBC), Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Tiago L Sampaio
- Laboratório de Bioprospecção Farmacêutica e Bioquímica Clínica (LBFBC), Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Alice Maria Costa Martins
- Laboratório de Bioprospecção Farmacêutica e Bioquímica Clínica (LBFBC), Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Edson H Teixeira
- Laboratório Integrado de Biomoléculas (LIBS), Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Ana Rafaela Freitas Dotto
- Programa de Pós-Graduação em Desenvolvimento Territorial Sustentável, Universidade Federal do Paraná, Matinhos, PR, Brazil
| | - Wanderlei do Amaral
- Departamento de Engenharia Química, Universidade Federal do Paraná, Curitiba, Paraná, Brasil
| | - Alexandre Magno R Teixeira
- Programa de Pós-Graduação em Biotecnologia - PPGB-Renorbio, Universidade Estadual do Ceará, Fortaleza, CE, Brazil.,Departamento de Química Biológica, Universidade Regional do Cariri, Crato, Brazil
| | - Pedro de Lima-Neto
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Emmanuel S Marinho
- Faculdade de Filosofia Dom Aureliano Matos, Universidade Estadual do Ceará, Limoeiro do Norte, Brazil
| | - Hélcio S Dos Santos
- Programa de Pós-Graduação em Biotecnologia - PPGB-Renorbio, Universidade Estadual do Ceará, Fortaleza, CE, Brazil.,Centro de Ciencias Exatas e Tecnologia, Universidade Estadual do Vale do Acaraú, Sobral, Brazil.,Departamento de Química Biológica, Universidade Regional do Cariri, Crato, Brazil
| |
Collapse
|
15
|
Ghosh AK, Yuan H. Total Syntheses of the Proposed Structure of Iriomoteolide-1a, -1b and Synthesis of Three Derivatives for Structural Studies. Mar Drugs 2022; 20:587. [PMID: 36286411 PMCID: PMC9605196 DOI: 10.3390/md20100587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/10/2023] Open
Abstract
Iriomoteolide-1a and iriomoteolide-1b are very potent cytotoxic agents, isolated from marine dinoflagellates. We carried out the enantioselective syntheses of the proposed structures of these natural products. However, our analysis of the NMR spectra of the synthetic iriomoteolide-1a and the natural products revealed that the structures of iriomoteolide-1a and iriomoteolide-1b were assigned incorrectly. Based upon our detailed analysis of the spectral data of the synthetic iriomoteolide-1a and the natural products, we rationally designed three diastereomers of the proposed structure of 1 in an effort to assign the correct structures. The key steps of our syntheses of the proposed structures of iriomoteolides involved a highly diastereoselective ene reaction, a carbocupration that utilized a Gilman reagent, a Julia-Kocienski olefination to couple fragments, and Yamaguchi macrolactonization to form the target macrolactone. This synthetic route was then utilized to carry out syntheses of three diastereomers to the proposed structure of 1. These diastereomeric structures show close similarities to natural iriomoteolide-1a; however, there were differences in their spectral data. While natural iriomoteolides exhibited potent cytotoxicies, our preliminary biological evaluation of synthetic iriomoteolide-1a, iriomoteolide-1b, and all three synthetic derivatives did not show any appreciable cytotoxic properties.
Collapse
Affiliation(s)
- Arun K. Ghosh
- Department of Chemistry, Department of Medicinal Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Hao Yuan
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
16
|
Li Y, Cheng S, Tian Y, Zhang Y, Zhao Y. Recent ring distortion reactions for diversifying complex natural products. Nat Prod Rep 2022; 39:1970-1992. [PMID: 35972343 DOI: 10.1039/d2np00027j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: 2013-2022.Chemical diversification of natural products is an efficient way to generate natural product-like compounds for modern drug discovery programs. Utilizing ring-distortion reactions for diversifying natural products would directly alter the core ring systems of small molecules and lead to the production of structurally complex and diverse compounds for high-throughput screening. We review the ring distortion reactions recently used in complexity-to-diversity (CtD) and pseudo natural products (pseudo-NPs) strategies for diversifying complex natural products. The core ring structures of natural products are altered via ring expansion, ring cleavage, ring edge-fusion, ring spiro-fusion, ring rearrangement, and ring contraction. These reactions can rapidly provide natural product-like collections with properties suitable for a wide variety of biological and medicinal applications. The challenges and limitations of current ring distortion reactions are critically assessed, and avenues for future improvements of this rapidly expanding field are discussed. We also provide a toolbox for chemists for the application of ring distortion reactions to access natural product-like molecules.
Collapse
Affiliation(s)
- Yu Li
- School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Shihao Cheng
- School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Yun Tian
- School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Yanan Zhang
- School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Yu Zhao
- School of Pharmacy, Nantong University, Nantong 226001, China.
| |
Collapse
|
17
|
Romanelli M, Amaral M, Thevenard F, Santa Cruz LM, Regasini LO, Migotto AE, Lago JHG, Tempone AG. Mitochondrial Imbalance of Trypanosoma cruzi Induced by the Marine Alkaloid 6-Bromo-2'-de- N-Methylaplysinopsin. ACS OMEGA 2022; 7:28561-28570. [PMID: 35990437 PMCID: PMC9387129 DOI: 10.1021/acsomega.2c03395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/27/2022] [Indexed: 06/09/2023]
Abstract
Chagas disease, caused by Trypanosoma cruzi, affects seven million people worldwide and lacks effective treatments. Using bioactivity-guided fractionation, NMR, and electrospray ionization-high resolution mass spectrometry (ESI-HRMS) spectral analysis, the indole alkaloid 6-bromo-2'-de-N-methylaplysinopsin (BMA) was isolated and chemically characterized from the marine coral Tubastraea tagusensis. BMA was tested against trypomastigotes and intracellular amastigotes of T. cruzi, resulting in IC50 values of 62 and 5.7 μM, respectively, with no mammalian cytotoxicity. The mechanism of action studies showed that BMA induced no alterations in the plasma membrane permeability but caused depolarization of the mitochondrial membrane potential, reducing ATP levels. Intracellular calcium levels were also reduced after the treatment, which was associated with pH alteration of acidocalcisomes. Using matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF)/MS analysis, alterations of mass spectral signals were observed after treatment with BMA, suggesting a different mechanism from benznidazole. In silico pharmacokinetic-pharmacodynamic (PKPD) parameters suggested a drug-likeness property, supporting the promising usefulness of this compound as a new hit for optimizations.
Collapse
Affiliation(s)
- Maiara
M. Romanelli
- Centre
for Parasitology and Mycology, Adolfo Lutz
Institute, Av Dr Arnaldo 351, São Paulo, SP 01246-000, Brazil
| | - Maiara Amaral
- Centre
for Parasitology and Mycology, Adolfo Lutz
Institute, Av Dr Arnaldo 351, São Paulo, SP 01246-000, Brazil
| | - Fernanda Thevenard
- Centre
of Natural Sciences and Humanities, Federal
University of ABC (UFABC), Avenida dos Estados 5001, Santo Andre, SP 09210-580, Brazil
| | - Lucas M. Santa Cruz
- Department
of Organic Contaminants, Instituto Adolfo
Lutz, Av Dr Arnaldo 355, São Paulo, SP 01246-000, Brazil
| | - Luis O. Regasini
- Department
of Chemistry and Environmental Sciences, Institute of Biosciences,
Humanities and Exact Sciences, Universidade
Estadual Paulista, R. Cristóvão Colombo 2265, São
Jose do Rio Preto, SP 15054-000, Brazil
| | - Alvaro E. Migotto
- Centre
for Marine Biology, Universidade de São
Paulo, Rodovia Manoel Hypólito do Rego, Km 131, São Sebastião, São Paulo, SP 11600-000, Brazil
| | - João Henrique G. Lago
- Centre
of Natural Sciences and Humanities, Federal
University of ABC (UFABC), Avenida dos Estados 5001, Santo Andre, SP 09210-580, Brazil
| | - Andre G. Tempone
- Centre
for Parasitology and Mycology, Adolfo Lutz
Institute, Av Dr Arnaldo 351, São Paulo, SP 01246-000, Brazil
| |
Collapse
|
18
|
Lu D, Zhang NZ, Yao Y, Wang T, Hua Q, Zheng X, Cong W, Tan F. Investigation of Antiparasitic Activity of Two Marine Natural Products, Estradiol Benzoate, and Octyl Gallate, on Toxoplasma gondii In Vitro. Front Pharmacol 2022; 13:841941. [PMID: 35370702 PMCID: PMC8968875 DOI: 10.3389/fphar.2022.841941] [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: 12/31/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Toxoplasmosis, caused by Toxoplasma gondii, is a common disease worldwide and could be severe and even fatal in immunocompromised individuals and fetuses. Limitation in current available treatment options drives the need to develop novel therapeutics. This study assessed the anti-T. gondii potential of 103 marine natural products. A luminescence-based β-galactosidase activity assay was used to screen the marine natural products library. Afterward, those compounds that displayed over 70% parasite inhibition ratio were further chosen to assess their cytotoxicity. Compounds exhibiting low cytotoxicity (≥80% cell viability) were applied to evaluate the inhibition efficacy on discrete steps of the T. gondii lytic cycle, including invasion, intracellular growth, and egress abilities as well as the cell cycle. We found that both estradiol benzoate and octyl gallate caused >70% inhibition of tachyzoite growth with IC50 values of 4.41 ± 0.94 and 5.66 ± 0.35 μM, respectively, and displayed low cytotoxicity with TD50 values of 34.11 ± 2.86 and 26.4 ± 0.98 μM, respectively. Despite their defects in inhibition of invasion and egress of tachyzoite, the two compounds markedly inhibited the tachyzoite intracellular replication. Flow cytometric analyses further suggested that the anti-T. gondii activity of estradiol benzoate, rather than octyl gallate, may be linked to halting cell cycle progression of tachyzoite from G1 to S phase. Taken together, these findings suggest that both estradiol benzoate and octyl gallate are potential inhibitors for anti-T. gondii infection and support the further exploration of marine natural products as a thinkable source of alternative and active agents against T. gondii.
Collapse
Affiliation(s)
- Daiqiang Lu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Nian-Zhang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Animal Echinococcosis Para-Reference Laboratory, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, China
| | - Yinning Yao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qianqian Hua
- Clinical Laboratory, Dongyang People's Hospital, Jinhua, China
| | - Xiaozi Zheng
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Cong
- Marine College, Shandong University, Weihai, China
| | - Feng Tan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Alonso-Vega C, Urbina JA, Sanz S, Pinazo MJ, Pinto JJ, Gonzalez VR, Rojas G, Ortiz L, Garcia W, Lozano D, Soy D, Maldonado RA, Nagarkatti R, Debrabant A, Schijman A, Thomas MC, López MC, Michael K, Ribeiro I, Gascon J, Torrico F, Almeida IC. New chemotherapy regimens and biomarkers for Chagas disease: the rationale and design of the TESEO study, an open-label, randomised, prospective, phase-2 clinical trial in the Plurinational State of Bolivia. BMJ Open 2021; 11:e052897. [PMID: 34972765 PMCID: PMC8720984 DOI: 10.1136/bmjopen-2021-052897] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Chagas disease (CD) affects ~7 million people worldwide. Benznidazole (BZN) and nifurtimox (NFX) are the only approved drugs for CD chemotherapy. Although both drugs are highly effective in acute and paediatric infections, their efficacy in adults with chronic CD (CCD) is lower and variable. Moreover, the high incidence of adverse events (AEs) with both drugs has hampered their widespread use. Trials in CCD adults showed that quantitative PCR (qPCR) assays remain negative for 12 months after standard-of-care (SoC) BZN treatment in ~80% patients. BZN pharmacokinetic data and the nonsynchronous nature of the proliferative mammal-dwelling parasite stage suggested that a lower BZN/NFX dosing frequency, combined with standard or extended treatment duration, might have the same or better efficacy than either drug SoC, with fewer AEs. METHODS AND ANALYSIS New ThErapies and Biomarkers for ChagaS infEctiOn (TESEO) is an open-label, randomised, prospective, phase-2 clinical trial, with six treatment arms (75 patients/arm, 450 patients). Primary objectives are to compare the safety and efficacy of two new proposed chemotherapy regimens of BZN and NFX in adults with CCD with the current SoC for BZN and NFX, evaluated by qPCR and biomarkers for 36 months posttreatment and correlated with CD conventional serology. Recruitment of patients was initiated on 18 December 2019 and on 20 May 2021, 450 patients (study goal) were randomised among the six treatment arms. The treatment phase was finalised on 18 August 2021. Secondary objectives include evaluation of population pharmacokinetics of both drugs in all treatment arms, the incidence of AEs, and parasite genotyping. ETHICS AND DISSEMINATION The TESEO study was approved by the National Institutes of Health (NIH), U.S. Food and Drug Administration (FDA), federal regulatory agency of the Plurinational State of Bolivia and the Ethics Committees of the participating institutions. The results will be disseminated via publications in peer-reviewed journals, conferences and reports to the NIH, FDA and participating institutions. TRIAL REGISTRATION NUMBER NCT03981523.
Collapse
Affiliation(s)
| | - Julio A Urbina
- Center for Biochemistry and Biophysics, Venezuelan Institute for Scientific Research (IVIC), Caracas, Distrito Capital, Venezuela, Bolivarian Republic of
| | - Sergi Sanz
- Biostatistics and Data Management Unit, Barcelona Institute for Global Health, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Basic Clinical Practice, Universitat de Barcelona, Barcelona, Spain
| | - María-Jesús Pinazo
- Barcelona Institute for Global Health (ISGLOBAL), Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Jimy José Pinto
- Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia, Plurinational State of
| | - Virginia R Gonzalez
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, Texas, USA
| | - Gimena Rojas
- Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia, Plurinational State of
| | - Lourdes Ortiz
- Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Tarija, Bolivia, Plurinational State of
- Universidad Autónoma Juan Misael Saracho, Tarija, Bolivia, Plurinational State of
| | - Wilson Garcia
- Centro Plataforma Chagas Sucre, Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Sucre, Bolivia, Plurinational State of
- Programa Departamental de Chagas Chuquisaca, Servicio Departamental de Salud de Chuquisaca, Chuquisaca, Bolivia, Plurinational State of
| | - Daniel Lozano
- Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia, Plurinational State of
| | - Dolors Soy
- Pharmacy Service, Division of Medicines, Hospital Clinic de Barcelona, Barcelona, Spain
- Institut de Investigació Biomèdica Agustí Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Rosa A Maldonado
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, Texas, USA
| | - Rana Nagarkatti
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Alain Debrabant
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Alejandro Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - M Carmen Thomas
- Consejo Superior de Investigaciones Científicas, Instituto de Parasitología y Biomedicina López-Neyra, Granada, Spain
| | - Manuel Carlos López
- Consejo Superior de Investigaciones Científicas, Instituto de Parasitología y Biomedicina López-Neyra, Granada, Spain
| | - Katja Michael
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas, USA
| | - Isabela Ribeiro
- Dynamic Portfolio Unit, Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGLOBAL), Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Faustino Torrico
- Fundación Ciencia y Estudios Aplicados para el Desarrollo en Salud y Medio Ambiente (CEADES), Cochabamba, Bolivia, Plurinational State of
| | - Igor C Almeida
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, Texas, USA
| |
Collapse
|
20
|
Endo T, Takemae H, Sharma I, Furuya T. Multipurpose Drugs Active Against Both Plasmodium spp. and Microorganisms: Potential Application for New Drug Development. Front Cell Infect Microbiol 2021; 11:797509. [PMID: 35004357 PMCID: PMC8740689 DOI: 10.3389/fcimb.2021.797509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/01/2021] [Indexed: 12/29/2022] Open
Abstract
Malaria, a disease caused by the protozoan parasites Plasmodium spp., is still causing serious problems in endemic regions in the world. Although the WHO recommends artemisinin combination therapies for the treatment of malaria patients, the emergence of artemisinin-resistant parasites has become a serious issue and underscores the need for the development of new antimalarial drugs. On the other hand, new and re-emergences of infectious diseases, such as the influenza pandemic, Ebola virus disease, and COVID-19, are urging the world to develop effective chemotherapeutic agents against the causative viruses, which are not achieved to the desired level yet. In this review article, we describe existing drugs which are active against both Plasmodium spp. and microorganisms including viruses, bacteria, and fungi. We also focus on the current knowledge about the mechanism of actions of these drugs. Our major aims of this article are to describe examples of drugs that kill both Plasmodium parasites and other microbes and to provide valuable information to help find new ideas for developing novel drugs, rather than merely augmenting already existing drug repurposing efforts.
Collapse
Affiliation(s)
- Takuro Endo
- Laboratory of Veterinary Infectious Diseases, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hitoshi Takemae
- Center for Infectious Disease Epidemiology and Prevention Research, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Indu Sharma
- Department of Biological Sciences, Hampton University, Hampton, VA, United States
| | - Tetsuya Furuya
- Laboratory of Veterinary Infectious Diseases, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
21
|
Tempone AG, Pieper P, Borborema SET, Thevenard F, Lago JHG, Croft SL, Anderson EA. Marine alkaloids as bioactive agents against protozoal neglected tropical diseases and malaria. Nat Prod Rep 2021; 38:2214-2235. [PMID: 34913053 PMCID: PMC8672869 DOI: 10.1039/d0np00078g] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Indexed: 01/09/2023]
Abstract
Covering: 2000 up to 2021Natural products are an important resource in drug discovery, directly or indirectly delivering numerous small molecules for potential development as human medicines. Among the many classes of natural products, alkaloids have a rich history of therapeutic applications. The extensive chemodiversity of alkaloids found in the marine environment has attracted considerable attention for such uses, while the scarcity of these natural materials has stimulated efforts towards their total synthesis. This review focuses on the biological activity of marine alkaloids (covering 2000 to up to 2021) towards Neglected Tropical Diseases (NTDs) caused by protozoan parasites, and malaria. Chemotherapy represents the only form of treatment for Chagas disease, human African trypanosomiasis, leishmaniasis and malaria, but there is currently a restricted arsenal of drugs, which often elicit severe adverse effects, show variable efficacy or resistance, or are costly. Natural product scaffolds have re-emerged as a focus of academic drug discovery programmes, offering a different resource to discover new chemical entities with new modes of action. In this review, the potential of a range of marine alkaloids is analyzed, accompanied by coverage of synthetic efforts that enable further studies of key antiprotozoal natural product scaffolds.
Collapse
Affiliation(s)
- Andre G Tempone
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, 01246-000, Brazil.
| | - Pauline Pieper
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| | - Samanta E T Borborema
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, 01246-000, Brazil.
| | - Fernanda Thevenard
- Centre of Natural Sciences and Humanities, Federal University of ABC, Sao Paulo, 09210-580, Brazil
| | - Joao Henrique G Lago
- Centre of Natural Sciences and Humanities, Federal University of ABC, Sao Paulo, 09210-580, Brazil
| | - Simon L Croft
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK.
| | - Edward A Anderson
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK.
| |
Collapse
|
22
|
Besednova NN, Zaporozhets TS, Andryukov BG, Kryzhanovsky SP, Ermakova SP, Kuznetsova TA, Voronova AN, Shchelkanov MY. Antiparasitic Effects of Sulfated Polysaccharides from Marine Hydrobionts. Mar Drugs 2021; 19:637. [PMID: 34822508 PMCID: PMC8624348 DOI: 10.3390/md19110637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
This review presents materials characterizing sulfated polysaccharides (SPS) of marine hydrobionts (algae and invertebrates) as potential means for the prevention and treatment of protozoa and helminthiasis. The authors have summarized the literature on the pathogenetic targets of protozoa on the host cells and on the antiparasitic potential of polysaccharides from red, brown and green algae as well as certain marine invertebrates. Information about the mechanisms of action of these unique compounds in diseases caused by protozoa has also been summarized. SPS is distinguished by high antiparasitic activity, good solubility and an almost complete absence of toxicity. In the long term, this allows for the consideration of these compounds as effective and attractive candidates on which to base drugs, biologically active food additives and functional food products with antiparasitic activity.
Collapse
Affiliation(s)
- Natalya N. Besednova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Tatyana S. Zaporozhets
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Boris G. Andryukov
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia
| | - Sergey P. Kryzhanovsky
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Svetlana P. Ermakova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Tatyana A. Kuznetsova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Anastasia N. Voronova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Mikhail Y. Shchelkanov
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia
- National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, 690041 Vladivostok, Russia
- Federal Scientific Center of the East Asia Terrestrial Biodiversity, Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia
| |
Collapse
|
23
|
Peres RB, Fiuza LFDA, da Silva PB, Batista MM, Camillo FDC, Marques AM, de C. Brito L, Figueiredo MR, Soeiro MDNC. In Vitro Phenotypic Activity and In Silico Analysis of Natural Products from Brazilian Biodiversity on Trypanosoma cruzi. Molecules 2021; 26:5676. [PMID: 34577145 PMCID: PMC8472459 DOI: 10.3390/molecules26185676] [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: 06/24/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chagas disease (CD) affects more than 6 million people worldwide. The available treatment is far from ideal, creating a demand for new alternative therapies. Botanical diversity provides a wide range of novel potential therapeutic scaffolds. Presently, our aim was to evaluate the mammalian host toxicity and anti-Trypanosoma cruzi activity of botanic natural products including extracts, fractions and purified compounds obtained from Brazilian flora. In this study, 36 samples of extracts and fractions and eight pure compounds obtained from seven plant species were evaluated. The fraction dichloromethane from Aureliana fasciculata var. fasciculata (AFfPD) and the crude extract of Piper tectoniifolium (PTFrE) showed promising trypanosomicidal activity. AFfPD and PTFrE presented EC50 values 10.7 ± 2.8 μg/mL and 12.85 ± 1.52 μg/mL against intracellular forms (Tulahuen strain), respectively. Additionally, both were active upon bloodstream trypomastigotes (Y strain), exhibiting EC50 2.2 ± 1.0 μg/mL and 38.8 ± 2.1 μg/mL for AFfPD and PTFrE, respectively. Importantly, AFfPD is about five-fold more potent than Benznidazole (Bz), the reference drug for CD, also reaching lower EC90 value (7.92 ± 2.2 μg/mL) as compared to Bz (23.3 ± 0.6 μg/mL). Besides, anti-parasitic effect of eight purified botanic substances was also investigated. Aurelianolide A and B (compounds 1 and 2) from A. fasciculata and compound 8 from P. tuberculatum displayed the best trypanosomicidal effect. Compounds 1, 2 and 8 showed EC50 of 4.6 ± 1.3 μM, 1.6 ± 0.4 μM and 8.1 ± 0.9 μM, respectively against intracellular forms. In addition, in silico analysis of these three biomolecules was performed to predict parameters of absorption, distribution, metabolism and excretion. The studied compounds presented similar ADMET profile as Bz, without presenting mutagenicity and hepatotoxicity aspects as predicted for Bz. Our findings indicate that these natural products have promising anti-T. cruzi effect and may represent new scaffolds for future lead optimization.
Collapse
Affiliation(s)
- Raiza B. Peres
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (R.B.P.); (L.F.d.A.F.); (P.B.d.S.); (M.M.B.)
| | - Ludmila F. de A. Fiuza
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (R.B.P.); (L.F.d.A.F.); (P.B.d.S.); (M.M.B.)
| | - Patrícia B. da Silva
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (R.B.P.); (L.F.d.A.F.); (P.B.d.S.); (M.M.B.)
| | - Marcos M. Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (R.B.P.); (L.F.d.A.F.); (P.B.d.S.); (M.M.B.)
| | - Flávia da C. Camillo
- Laboratório de Tecnologia para Biodiversidade em Saúde/LDFito, Instituto de Tecnologia em Fármacos (Farmanguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (F.d.C.C.); (A.M.M.); (L.d.C.B.); (M.R.F.)
| | - André M. Marques
- Laboratório de Tecnologia para Biodiversidade em Saúde/LDFito, Instituto de Tecnologia em Fármacos (Farmanguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (F.d.C.C.); (A.M.M.); (L.d.C.B.); (M.R.F.)
| | - Lavínia de C. Brito
- Laboratório de Tecnologia para Biodiversidade em Saúde/LDFito, Instituto de Tecnologia em Fármacos (Farmanguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (F.d.C.C.); (A.M.M.); (L.d.C.B.); (M.R.F.)
| | - Maria R. Figueiredo
- Laboratório de Tecnologia para Biodiversidade em Saúde/LDFito, Instituto de Tecnologia em Fármacos (Farmanguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (F.d.C.C.); (A.M.M.); (L.d.C.B.); (M.R.F.)
| | - Maria de N. C. Soeiro
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil 4365, Manguinhos, Rio de Janeiro 210360-040, Brazil; (R.B.P.); (L.F.d.A.F.); (P.B.d.S.); (M.M.B.)
| |
Collapse
|
24
|
Petersen KV, Selas A, Hymøller KM, Mizielinski K, Thorsager M, Stougaard M, Alonso C, Palacios F, Pérez-Pertejo Y, Reguera RM, Balaña-Fouce R, Knudsen BR, Tesauro C. Simple and Fast DNA Based Sensor System for Screening of Small-Molecule Compounds Targeting Eukaryotic Topoisomerase 1. Pharmaceutics 2021; 13:1255. [PMID: 34452216 PMCID: PMC8401307 DOI: 10.3390/pharmaceutics13081255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Eukaryotic topoisomerase 1 is a potential target of anti-parasitic and anti-cancer drugs. Parasites require topoisomerase 1 activity for survival and, consequently, compounds that inhibit topoisomerase 1 activity may be of interest. All effective topoisomerase 1 drugs with anti-cancer activity act by inhibiting the ligation reaction of the enzyme. Screening for topoisomerase 1 targeting drugs, therefore, should involve the possibility of dissecting which step of topoisomerase 1 activity is affected. Methods: Here we present a novel DNA-based assay that allows for screening of the effect of small-molecule compounds targeting the binding/cleavage or the ligation steps of topoisomerase 1 catalysis. This novel assay is based on the detection of a rolling circle amplification product generated from a DNA circle resulting from topoisomerase 1 activity. Results: We show that the binding/cleavage and ligation reactions of topoisomerase 1 can be investigated separately in the presented assay termed REEAD (C|L) and demonstrate that the assay can be used to investigate, which of the individual steps of topoisomerase 1 catalysis are affected by small-molecule compounds. The assay is gel-free and the results can be detected by a simple colorimetric readout method using silver-on-gold precipitation rendering large equipment unnecessary. Conclusion: REEAD (C|L) allows for easy and quantitative investigations of topoisomerase 1 targeting compounds and can be performed in non-specialized laboratories.
Collapse
Affiliation(s)
- Kamilla Vandsø Petersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (K.V.P.); (K.M.H.); (B.R.K.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Asier Selas
- Department of Organic Chemistry, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (A.S.); (C.A.); (F.P.)
| | - Kirstine Mejlstrup Hymøller
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (K.V.P.); (K.M.H.); (B.R.K.)
| | | | - Maria Thorsager
- VPCIR Biosciences ApS., 8000 Aarhus, Denmark; (K.M.); (M.T.)
| | - Magnus Stougaard
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
- VPCIR Biosciences ApS., 8000 Aarhus, Denmark; (K.M.); (M.T.)
- Department of Pathology, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Concepcion Alonso
- Department of Organic Chemistry, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (A.S.); (C.A.); (F.P.)
| | - Francisco Palacios
- Department of Organic Chemistry, University of Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (A.S.); (C.A.); (F.P.)
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences, University of Leon (ULE), 24071 Leon, Spain; (Y.P.-P.); (R.M.R.); (R.B.-F.)
| | - Rosa M. Reguera
- Department of Biomedical Sciences, University of Leon (ULE), 24071 Leon, Spain; (Y.P.-P.); (R.M.R.); (R.B.-F.)
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences, University of Leon (ULE), 24071 Leon, Spain; (Y.P.-P.); (R.M.R.); (R.B.-F.)
| | - Birgitta R. Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (K.V.P.); (K.M.H.); (B.R.K.)
- VPCIR Biosciences ApS., 8000 Aarhus, Denmark; (K.M.); (M.T.)
| | - Cinzia Tesauro
- VPCIR Biosciences ApS., 8000 Aarhus, Denmark; (K.M.); (M.T.)
| |
Collapse
|
25
|
Wright AE, Collins JE, Roberts B, Roberts JC, Winder PL, Reed JK, Diaz MC, Pomponi SA, Chakrabarti D. Antiplasmodial Compounds from Deep-Water Marine Invertebrates. Mar Drugs 2021; 19:md19040179. [PMID: 33805935 PMCID: PMC8064351 DOI: 10.3390/md19040179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 01/03/2023] Open
Abstract
Novel drug leads for malaria therapy are urgently needed because of the widespread emergence of resistance to all available drugs. Screening of the Harbor Branch enriched fraction library against the Plasmodium falciparum chloroquine-resistant strain (Dd2) followed by bioassay-guided fractionation led to the identification of two potent antiplasmodials; a novel diterpene designated as bebrycin A (1) and the known C21 degraded terpene nitenin (2). A SYBR Green I assay was used to establish a Dd2 EC50 of 1.08 ± 0.21 and 0.29 ± 0.02 µM for bebrycin A and nitenin, respectively. Further analysis was then performed to assess the stage specificity of the inhibitors antiplasmodial effects on the Dd2 intraerythrocytic life cycle. Exposure to bebrycin A was found to block parasite maturation at the schizont stage if added any time prior to late schizogony at 42 hours post invasion, (HPI). In contrast, early life cycle exposure to nitenin (prior to 18 HPI) was identified as crucial to parasite inhibition, suggesting nitenin may target the maturation of the parasite during the transition from ring to early trophozoite (6–18 HPI), a novel property among known antimalarials.
Collapse
Affiliation(s)
- Amy E. Wright
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
- Correspondence: (A.E.W.); (D.C.); Tel.: +1-772-242-2459 (A.E.W.); +1-407-882-2256 (D.C.)
| | - Jennifer E. Collins
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA; (J.E.C.); (B.R.)
| | - Bracken Roberts
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA; (J.E.C.); (B.R.)
| | - Jill C. Roberts
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
| | - Priscilla L. Winder
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
| | - John K. Reed
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
| | - Maria Cristina Diaz
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
| | - Shirley A. Pomponi
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US Highway 1 North, Fort Pierce, FL 34946, USA; (J.C.R.); (P.L.W.); (J.K.R.); (M.C.D.); (S.A.P.)
| | - Debopam Chakrabarti
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA; (J.E.C.); (B.R.)
- Correspondence: (A.E.W.); (D.C.); Tel.: +1-772-242-2459 (A.E.W.); +1-407-882-2256 (D.C.)
| |
Collapse
|
26
|
El-Demerdash A, Metwaly AM, Hassan A, Abd El-Aziz TM, Elkaeed EB, Eissa IH, Arafa RK, Stockand JD. Comprehensive Virtual Screening of the Antiviral Potentialities of Marine Polycyclic Guanidine Alkaloids against SARS-CoV-2 (COVID-19). Biomolecules 2021; 11:460. [PMID: 33808721 PMCID: PMC8003478 DOI: 10.3390/biom11030460] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
The huge global expansion of the COVID-19 pandemic caused by the novel SARS-corona virus-2 is an extraordinary public health emergency. The unavailability of specific treatment against SARS-CoV-2 infection necessitates the focus of all scientists in this direction. The reported antiviral activities of guanidine alkaloids encouraged us to run a comprehensive in silico binding affinity of fifteen guanidine alkaloids against five different proteins of SARS-CoV-2, which we investigated. The investigated proteins are COVID-19 main protease (Mpro) (PDB ID: 6lu7), spike glycoprotein (PDB ID: 6VYB), nucleocapsid phosphoprotein (PDB ID: 6VYO), membrane glycoprotein (PDB ID: 6M17), and a non-structural protein (nsp10) (PDB ID: 6W4H). The binding energies for all tested compounds indicated promising binding affinities. A noticeable superiority for the pentacyclic alkaloids particularly, crambescidin 786 (5) and crambescidin 826 (13) has been observed. Compound 5 exhibited very good binding affinities against Mpro (ΔG = -8.05 kcal/mol), nucleocapsid phosphoprotein (ΔG = -6.49 kcal/mol), and nsp10 (ΔG = -9.06 kcal/mol). Compound 13 showed promising binding affinities against Mpro (ΔG = -7.99 kcal/mol), spike glycoproteins (ΔG = -6.95 kcal/mol), and nucleocapsid phosphoprotein (ΔG = -8.01 kcal/mol). Such promising activities might be attributed to the long ω-fatty acid chain, which may play a vital role in binding within the active sites. The correlation of c Log P with free binding energies has been calculated. Furthermore, the SAR of the active compounds has been clarified. The Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) studies were carried out in silico for the 15 compounds; most examined compounds showed optimal to good range levels of ADMET aqueous solubility, intestinal absorption and being unable to pass blood brain barrier (BBB), non-inhibitors of CYP2D6, non-hepatotoxic, and bind plasma protein with a percentage less than 90%. The toxicity of the tested compounds was screened in silico against five models (FDA rodent carcinogenicity, carcinogenic potency TD50, rat maximum tolerated dose, rat oral LD50, and rat chronic lowest observed adverse effect level (LOAEL)). All compounds showed expected low toxicity against the tested models. Molecular dynamic (MD) simulations were also carried out to confirm the stable binding interactions of the most promising compounds, 5 and 13, with their targets. In conclusion, the examined 15 alkaloids specially 5 and 13 showed promising docking, ADMET, toxicity and MD results which open the door for further investigations for them against SARS-CoV-2.
Collapse
Affiliation(s)
- Amr El-Demerdash
- Metabolic Biology & Biological Chemistry Department, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
- Organic Chemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed M. Metwaly
- Department of Pharmacognosy & Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Afnan Hassan
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza 12578, Egypt; (A.H.); (R.K.A.)
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA;
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Riyadh, Saudi Arabia;
| | - Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt;
| | - Reem K. Arafa
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Giza 12578, Egypt; (A.H.); (R.K.A.)
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 12578, Egypt
| | - James D. Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA;
| |
Collapse
|
27
|
Chan-Bacab MJ, Reyes-Estebanez MM, Camacho-Chab JC, Ortega-Morales BO. Microorganisms as a Potential Source of Molecules to Control Trypanosomatid Diseases. Molecules 2021; 26:molecules26051388. [PMID: 33806654 PMCID: PMC7962016 DOI: 10.3390/molecules26051388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 11/17/2022] Open
Abstract
Trypanosomatids are the causative agents of leishmaniasis and trypanosomiasis, which affect about 20 million people in the world’s poorest countries, leading to 95,000 deaths per year. They are often associated with malnutrition, weak immune systems, low quality housing, and population migration. They are generally recognized as neglected tropical diseases. New drugs against these parasitic protozoa are urgently needed to counteract drug resistance, toxicity, and the high cost of commercially available drugs. Microbial bioprospecting for new molecules may play a crucial role in developing a new generation of antiparasitic drugs. This article reviews the current state of the available literature on chemically defined metabolites of microbial origin that have demonstrated antitrypanosomatid activity. In this review, bacterial and fungal metabolites are presented; they originate from a range of microorganisms, including cyanobacteria, heterotrophic bacteria, and filamentous fungi. We hope to provide a useful overview for future research to identify hits that may become the lead compounds needed to accelerate the discovery of new drugs against trypanosomatids.
Collapse
|
28
|
Nweze JA, Mbaoji FN, Li YM, Yang LY, Huang SS, Chigor VN, Eze EA, Pan LX, Zhang T, Yang DF. Potentials of marine natural products against malaria, leishmaniasis, and trypanosomiasis parasites: a review of recent articles. Infect Dis Poverty 2021; 10:9. [PMID: 33482912 PMCID: PMC7821695 DOI: 10.1186/s40249-021-00796-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/06/2021] [Indexed: 12/26/2022] Open
Abstract
Background Malaria and neglected communicable protozoa parasitic diseases, such as leishmaniasis, and trypanosomiasis, are among the otherwise called diseases for neglected communities, which are habitual in underprivileged populations in developing tropical and subtropical regions of Africa, Asia, and the Americas. Some of the currently available therapeutic drugs have some limitations such as toxicity and questionable efficacy and long treatment period, which have encouraged resistance. These have prompted many researchers to focus on finding new drugs that are safe, effective, and affordable from marine environments. The aim of this review was to show the diversity, structural scaffolds, in-vitro or in-vivo efficacy, and recent progress made in the discovery/isolation of marine natural products (MNPs) with potent bioactivity against malaria, leishmaniasis, and trypanosomiasis. Main text We searched PubMed and Google scholar using Boolean Operators (AND, OR, and NOT) and the combination of related terms for articles on marine natural products (MNPs) discovery published only in English language from January 2016 to June 2020. Twenty nine articles reported the isolation, identification and antiparasitic activity of the isolated compounds from marine environment. A total of 125 compounds were reported to have been isolated, out of which 45 were newly isolated compounds. These compounds were all isolated from bacteria, a fungus, sponges, algae, a bryozoan, cnidarians and soft corals. In recent years, great progress is being made on anti-malarial drug discovery from marine organisms with the isolation of these potent compounds. Comparably, some of these promising antikinetoplastid MNPs have potency better or similar to conventional drugs and could be developed as both antileishmanial and antitrypanosomal drugs. However, very few of these MNPs have a pharmaceutical destiny due to lack of the following: sustainable production of the bioactive compounds, standard efficient screening methods, knowledge of the mechanism of action, partnerships between researchers and pharmaceutical industries. Conclusions It is crystal clear that marine organisms are a rich source of antiparasitic compounds, such as alkaloids, terpenoids, peptides, polyketides, terpene, coumarins, steroids, fatty acid derivatives, and lactones. The current and future technological innovation in natural products drug discovery will bolster the drug armamentarium for malaria and neglected tropical diseases.
Collapse
Affiliation(s)
- Justus Amuche Nweze
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China.,Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria.,Department of Science Laboratory Technology, Faculty of Physical Sciences, University of Nigeria, Nsukka, Nigeria.,College of Life Science and Technology of Guangxi University, Nanning, 530004, Guangxi, People's Republic of China
| | - Florence N Mbaoji
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China.,College of Life Science and Technology of Guangxi University, Nanning, 530004, Guangxi, People's Republic of China.,Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Nigeria
| | - Yan-Ming Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China
| | - Li-Yan Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China
| | - Shu-Shi Huang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China
| | - Vincent N Chigor
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria.,Water and Public Health Research Group, University of Nigeria, Nsukka, PMB 410001, Enugu State, Nigeria
| | - Emmanuel A Eze
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Li-Xia Pan
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China
| | - Ting Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, 200025, People's Republic of China. .,National Health Commission Key Laboratory of Echinococcosis Prevention and Control, Xizang Center for Disease Control and Prevention, Linlang North Road, Lhasa, 850000, Tibet Autonomous Region, People's Republic of China.
| | - Deng-Feng Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, Guangxi, People's Republic of China.
| |
Collapse
|
29
|
Akao Y, Canan S, Cao Y, Condroski K, Engkvist O, Itono S, Kaki R, Kimura C, Kogej T, Nagaoka K, Naito A, Nakai H, Pairaudeau G, Radu C, Roberts I, Shimada M, Shum D, Watanabe NA, Xie H, Yonezawa S, Yoshida O, Yoshida R, Mowbray C, Perry B. Collaborative virtual screening to elaborate an imidazo[1,2- a]pyridine hit series for visceral leishmaniasis. RSC Med Chem 2021; 12:384-393. [PMID: 34041487 PMCID: PMC8130605 DOI: 10.1039/d0md00353k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
An innovative pre-competitive virtual screening collaboration was engaged to validate and subsequently explore an imidazo[1,2-a]pyridine screening hit for visceral leishmaniasis. In silico probing of five proprietary pharmaceutical company libraries enabled rapid expansion of the hit chemotype, alleviating initial concerns about the core chemical structure while simultaneously improving antiparasitic activity and selectivity index relative to the background cell line. Subsequent hit optimization informed by the structure–activity relationship enabled by this virtual screening allowed thorough investigation of the pharmacophore, opening avenues for further improvement and optimization of the chemical series. Ligand-based similarity screening of proprietary pharmaceutical company libraries enables rapid hit to lead investigation of a chemotype with anti-leishmania activity.![]()
Collapse
Affiliation(s)
- Yuichiro Akao
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - Stacie Canan
- Celgene Corporation, Celgene Global Health 10300 Campus Point Drive San Diego California 92121 USA
| | - Yafeng Cao
- WuXi AppTec Company Ltd. 666 Gaoxin Road, East Lake High-Tech Development Zone Wuhan 430075 People's Republic of China
| | - Kevin Condroski
- Celgene Corporation, Celgene Global Health 10300 Campus Point Drive San Diego California 92121 USA
| | - Ola Engkvist
- AstraZeneca Discovery Sciences, R&D AstraZeneca Gothenburg Sweden
| | - Sachiko Itono
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - Rina Kaki
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Chiaki Kimura
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Thierry Kogej
- AstraZeneca Discovery Sciences, R&D AstraZeneca Gothenburg Sweden
| | - Kazuya Nagaoka
- Eisai Co., Ltd 1-3,Tokodai 5-chome Tsukuba Ibaraki 300-2635 Japan
| | - Akira Naito
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Hiromi Nakai
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | | | - Constantin Radu
- Institut Pasteur Korea 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu Seongnam-si Gyeonggi-do 13488 Republic of Korea
| | - Ieuan Roberts
- AstraZeneca, Discovery Sciences, R&D AstraZeneca Cambridge UK
| | - Mitsuyuki Shimada
- Takeda Pharmaceutical Company Limited 26-1 Muraoka-Higashi 2-chrome Fujisawa Kanagawa 251-8555 Japan
| | - David Shum
- Institut Pasteur Korea 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu Seongnam-si Gyeonggi-do 13488 Republic of Korea
| | - Nao-Aki Watanabe
- Eisai Co., Ltd 1-3,Tokodai 5-chome Tsukuba Ibaraki 300-2635 Japan
| | - Huanxu Xie
- WuXi AppTec Company Ltd. 666 Gaoxin Road, East Lake High-Tech Development Zone Wuhan 430075 People's Republic of China
| | - Shuji Yonezawa
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Osamu Yoshida
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Ryu Yoshida
- Shionogi & Co., Ltd 3-1-1, Futaba-cho Toyonaka-shi Osaka Japan
| | - Charles Mowbray
- Drugs for Neglected Diseases initiative 15 Chemin Louis Dunant Geneva 1202 Switzerland
| | - Benjamin Perry
- Drugs for Neglected Diseases initiative 15 Chemin Louis Dunant Geneva 1202 Switzerland
| |
Collapse
|
30
|
Sánchez-Suárez J, Bernal FA, Coy-Barrera E. Colombian Contributions Fighting Leishmaniasis: A Systematic Review on Antileishmanials Combined with Chemoinformatics Analysis. Molecules 2020; 25:E5704. [PMID: 33287235 PMCID: PMC7730898 DOI: 10.3390/molecules25235704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 01/15/2023] Open
Abstract
Leishmaniasis is a parasitic morbid/fatal disease caused by Leishmania protozoa. Twelve million people worldwide are appraised to be currently infected, including ca. two million infections each year, and 350 million people in 88 countries are at risk of becoming infected. In Colombia, cutaneous leishmaniasis (CL) is a public health problem in some tropical areas. Therapeutics is based on traditional antileishmanial drugs, but this practice has several drawbacks for patients. Thus, the search for new antileishmanial agents is a serious need, but the lack of adequately funded research programs on drug discovery has hampered its progress. Some Colombian researchers have conducted different research projects focused on the assessment of the antileishmanial activity of naturally occurring and synthetic compounds against promastigotes and/or amastigotes. Results of such studies have separately demonstrated important hits and reasonable potential, but a holistic view of them is lacking. Hence, we present the outcome from a systematic review of the literature (under PRISMA guidelines) on those Colombian studies investigating antileishmanials during the last thirty-two years. In order to combine the general efforts aiming at finding a lead against Leishmania panamensis (one of the most studied and incident parasites in Colombia causing CL) and to recognize structural features of representative compounds, fingerprint-based analyses using conventional machine learning algorithms and clustering methods are shown. Abstraction from such a meta-description led to describe some function-determining molecular features and simplify the clustering of plausible isofunctional hits. This systematic review indicated that the Colombian efforts for the antileishmanials discovery are increasingly intensified, though improvements in the followed pathways must be definitively pursued. In this context, a brief discussion about scope, strengths and limitations of such advances and relationships is addressed.
Collapse
Affiliation(s)
- Jeysson Sánchez-Suárez
- Bioprospecting Research Group, School of Engineering, Universidad de La Sabana, Chía 250001, Colombia;
| | - Freddy A. Bernal
- Bioorganic Chemistry Laboratory, Universidad Militar Nueva Granada, Cajicá 250247, Colombia;
| | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Universidad Militar Nueva Granada, Cajicá 250247, Colombia;
| |
Collapse
|
31
|
Mansoldo FRP, Carta F, Angeli A, Cardoso VDS, Supuran CT, Vermelho AB. Chagas Disease: Perspectives on the Past and Present and Challenges in Drug Discovery. Molecules 2020; 25:E5483. [PMID: 33238613 PMCID: PMC7700143 DOI: 10.3390/molecules25225483] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease still has no effective treatment option for all of its phases despite being discovered more than 100 years ago. The development of commercial drugs has been stagnating since the 1960s, a fact that sheds light on the question of how drug discovery research has progressed and taken advantage of technological advances. Could it be that technological advances have not yet been sufficient to resolve this issue or is there a lack of protocol, validation and standardization of the data generated by different research teams? This work presents an overview of commercial drugs and those that have been evaluated in studies and clinical trials so far. A brief review is made of recent target-based and phenotypic studies based on the search for molecules with anti-Trypanosoma cruzi action. It also discusses how proteochemometric (PCM) modeling and microcrystal electron diffraction (MicroED) can help in the case of the lack of a 3D protein structure; more specifically, Trypanosoma cruzi carbonic anhydrase.
Collapse
Affiliation(s)
- Felipe Raposo Passos Mansoldo
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| | - Fabrizio Carta
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
| | - Andrea Angeli
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania
| | - Veronica da Silva Cardoso
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| | - Claudiu T. Supuran
- Neurofarba Department, Università degli Studi di Firenze, Sezione di Scienze Farmaceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy; (F.C.); (A.A.)
| | - Alane Beatriz Vermelho
- BIOINOVAR-Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (F.R.P.M.); (V.d.S.C.)
| |
Collapse
|
32
|
Bailly C. Pyronaridine: An update of its pharmacological activities and mechanisms of action. Biopolymers 2020; 112:e23398. [PMID: 33280083 DOI: 10.1002/bip.23398] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Pyronaridine (PYR) is an erythrocytic schizonticide with a potent antimalarial activity against multidrug-resistant Plasmodium. The drug is used in combination with artesunate for the treatment of uncomplicated P. falciparum malaria, in adults and children. The present review briefly retraces the discovery of PYR and recent antimalarial studies which has led to the approval of PYR/artesunate combination (Pyramax) by the European Medicines Agency to treat uncomplicated malaria worldwide. PYR also presents a marked antitumor activity and has revealed efficacy for the treatment of other parasitic diseases (notably Babesia and Trypanosoma infections) and to mitigate the Ebola virus propagation. On the one hand, PYR functions has an inhibitor of hemozoin (biomineral malaria pigment, by-product of hemoglobin digestion) formation, blocking the biopolymerization of β-hematin and thus facilitating the accumulation of toxic hematin into the digestive vacuole of the parasite. On the other hand, PYR is a bona fide DNA-intercalating agent and an inhibitor of DNA topoisomerase 2, leading to DNA damages and cell death. Inhibition of hematin polymerization represents the prime mechanism at the origin of the antimalarial activity, whereas anticancer effects relies essentially on the interference with DNA metabolism, as with structurally related anticancer drugs like amsacrine and quinacrine. In addition, recent studies point to an immune modulatory activity of PYR and the implication of a mitochondrial oxidative pathway. An analogy with the mechanism of action of artemisinin drugs is underlined. In brief, the biological actions of pyronaridine are recapitulated to shed light on the diverse health benefits of this unsung drug.
Collapse
|