1
|
Mohammed MMD, Mohammed HS, El Wafa SAA, Ahmed DA, Heikal EA, Elgohary I, Barakat AM. Discovery of potent anti-toxoplasmosis drugs from secondary metabolites in Citrus limon (lemon) leaves, supported in-silico study. Sci Rep 2025; 15:624. [PMID: 39753625 PMCID: PMC11698829 DOI: 10.1038/s41598-024-82787-9] [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: 08/11/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
Toxoplasmosis induced by Toxoplasma gondii is a well-known health threat, that prompts fatal encephalitis increased with immunocompromised patients, in addition, it can cause chorioretinitis, microcephaly, stillbirth in the fetus and even led to death. Standard therapy uses sulfadiazine and pyrimethamine drugs revealed beneficial results during the acute stage, however, it has severe side effects. UPLC-ESI-MS/MS used to explore C. limon MeOH ext. constituents, which revealed a list of 41 metabolites of different classes encompasses; unsaturated fatty acid, tricarboxylic acids, phenolic aldehyde, phenolic acids, phenolic glycosides, coumarins, sesquiterpene lactone, limonoid, steroid and flavonoids. C. limon MeOH ext. and the isolates reduced significantly the number of T. gondii tachyzoites. Consequently, histopathological examination, proved significant reduction in the number of mononuclear inflammatory cells in the kidney and liver sections, besides, lowering the number of shrunken and degenerative neurons in the brain sections of infected mice. Molecular docking study was performed targeted certain receptors, which are important for the life cycle fundamentals for the parasite mobility including invasion and egress, and further molecular dynamics simulation was conducted to get insights into the structural changes of the formed complexes, along with a pharmacophoric mapping approach, that confirmed the need for a free hydroxyl group and/or a phenolic substituted one, in order to form HB, Hyd/Aro and ML interactions, through which, cell cycle disruption via iron chelation, could be achieved. In addition, the ADMIT properties of all identified metabolites were predicted.
Collapse
Affiliation(s)
- Magdy Mostafa Desoky Mohammed
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt.
| | - Hala Sh Mohammed
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| | - Salwa A Abu El Wafa
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Doaa A Ahmed
- Medical Parasitology Department, Faculty of Medicine, Al-Azhar University for Girls, Cairo, Egypt
| | - Elham A Heikal
- Medical Parasitology Department, Faculty of Medicine, Al-Azhar University for Girls, Cairo, Egypt
| | - Islam Elgohary
- Department of Pathology, Agriculture Research Centre, Animal Health Research Institute, Dokki, Giza, Egypt
| | - Ashraf M Barakat
- Department of Zoonotic Diseases, National Research Centre, Dokki, Giza, 12622, Egypt
| |
Collapse
|
2
|
Nguyen TD, Choi JA, Lim HJ, Chae CH, Lee J, Son SH, Kim J, Son D, Kim HJ, Song CH. Inhibitors of acetohydroxyacid synthase as promising agents against non-tuberculous mycobacterial diseases. J Antibiot (Tokyo) 2024:10.1038/s41429-024-00799-z. [PMID: 39672903 DOI: 10.1038/s41429-024-00799-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Acetohydroxyacid synthase (AHAS), exclusively present in microorganisms and plants, is a promising target for several herbicides due to its catalytic role in the branched-chain amino acid biosynthetic pathway. Previous studies have shown that K13787, a pyrazolopyrimidine sulfonamide AHAS inhibitor, was moderately effective against pulmonary infection caused by M. tuberculosis and nontuberculous mycobacteria (NTM). In this study, we synthesized various structural derivatives of K13787 based on the molecular docking studies and assessed their MICs against mycobacteria species. Among the synthetic compounds screened, K13787, along with KNT2077 and KNT2099, exhibited inhibitory efficacy against M. avium and M. abscessus, including CLR-resistant NTM species. Notably, these compounds displayed a synergistic effect (FIC ≤ 0.5) when combined with CLR against M. avium and M. abscessus. Our findings suggest that these newly identified AHAS-targeted compounds hold promise as lead candidates for novel antimycobacterial agents against NTM infections. Considering the structure-activity relationship, K13787, KNT2077, and KTN2099 emerge as potential treatments for NTM species.
Collapse
Affiliation(s)
- Tam Doan Nguyen
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Ji-Ae Choi
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Hee-Jong Lim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Chong Hak Chae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Junghwan Lee
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Sang-Hun Son
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Jaewhan Kim
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Doyi Son
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Translational Immunology Institute, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
| | - Chang-Hwa Song
- Department of Microbiology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Translational Immunology Institute, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
| |
Collapse
|
3
|
Liu S, Cai M, Liu Z, Gao W, Li J, Li Y, Abudouxukuer X, Zhang J. Comprehensive Insights into the Development of Antitoxoplasmosis Drugs: Current Advances, Obstacles, and Future Perspectives. J Med Chem 2024; 67:20740-20764. [PMID: 39589152 DOI: 10.1021/acs.jmedchem.4c01733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Current therapies for toxoplasmosis rely on a few drugs, most of which have severe side effects, and seeking ideal therapies for different types of toxoplasmosis is a long-term and challenging mission. Research and development (R&D) of novel drugs against Toxoplasma gondii (T. gondii) has focused on two main directions, the structural modification of lead compounds and natural products. Here we summarize the recent advances in the development of anti-T. gondii drugs from these two perspectives and provide comprehensive insights, reflecting on the advantages and selected molecules in each field. This review also focuses on the current obstacles to the development of novel anti-T. gondii agents, proposes comprehensive solutions, and facilitates future development.
Collapse
Affiliation(s)
- Siyang Liu
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Minghao Cai
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Zhendi Liu
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Weixin Gao
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Junjie Li
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Yuxueqing Li
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Xiayire Abudouxukuer
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| | - Jili Zhang
- Health Science Center, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 513211, China
| |
Collapse
|
4
|
Ariefta NR, Sofian FF, Aboshi T, Kuncoro H, Dinata DI, Shiono Y, Nishikawa Y. Evaluation of the antiplasmodial and anti-Toxoplasma activities of several Indonesian medicinal plant extracts. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118269. [PMID: 38697409 DOI: 10.1016/j.jep.2024.118269] [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: 02/28/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Malaria, caused by Plasmodium parasites, remains a significant global health challenge, particularly in tropical and subtropical regions. At the same time, the prevalence of toxoplasmosis has been reported to be 30% worldwide. Traditional medicines have long played a vital role in discovering and developing novel drugs, and this approach is essential in the face of increasing resistance to current antimalarial and anti-Toxoplasma drugs. In Indonesian traditional medicine, various plants are used for their therapeutic properties. This study focuses on eleven medicinal plants from which nineteen extracts were obtained and screened for their potential medicinal benefits against malaria and toxoplasmosis. AIMS OF THE STUDY The aim of this study was to evaluate the efficacy of extracts from Indonesian medicinal plants to inhibit Plasmodium falciparum, a parasite responsible for malaria, and Toxoplasma gondii, an opportunistic parasite responsible for toxoplasmosis. METHODS Nineteen extracts from eleven plants were subjected to in vitro screening against P. falciparum 3D7 (a chloroquine-sensitive strain) and the T. gondii RH strain. In vitro treatments were conducted on P. falciparum 3D7 and K1 (multidrug-resistant strains) using the potent extracts, and in vivo assessments were carried out with mice infected with P. yoelii 17XNL. LCMS analysis was also conducted to identify the main components of the most effective extract. RESULTS Seven extracts showed significant antiplasmodial activity (>80% inhibition) at a concentration of 100 μg/ml. These extracts were obtained from Dysoxylum parasiticum (Osbeck) Kosterm., Elaeocarpus glaber (Bl.) Bijdr., Eleutherine americana Merr., Kleinhovia hospita L., Peronema canescens Jack, and Plectranthus scutellarioides (L.) R.Br. Notably, the D. parasiticum ethyl acetate extract exhibited high selectivity and efficacy both in vitro and in vivo. Herein, the key active compounds oleamide and erucamide were identified, which had IC50 values (P. falciparum 3D7/K1) of 17.49/23.63 μM and 32.49/51.59 μM, respectively. CONCLUSIONS The results of this study highlight the antimalarial potential of plant extracts collected from Indonesia. Particularly, extracts from D. parasiticum EtOH and EtOAc stood out for their low toxicity and strong antiplasmodial properties, with the EtOAc extract emerging as a notably promising antimalarial candidate. Key compounds identified within this extract demonstrate the complexity of extracts' action against malaria, potentially targeting both the parasite and the host. This suggests a promising approach for developing new antimalarial strategies that tackle the multifaceted challenges of drug resistance and disease management. Future investigations are necessary to unlock the full therapeutic potential of these extracts.
Collapse
Affiliation(s)
- Nanang Rudianto Ariefta
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| | - Ferry Ferdiansyah Sofian
- Department of Life, Food, and Environmental Sciences, Faculty of Agriculture, Yamagata University, Wakaba-machi 1-23, Tsuruoka, Yamagata, 997-8555, Japan; Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Sumedang, West Java, 45363, Indonesia.
| | - Takako Aboshi
- Department of Life, Food, and Environmental Sciences, Faculty of Agriculture, Yamagata University, Wakaba-machi 1-23, Tsuruoka, Yamagata, 997-8555, Japan.
| | - Hadi Kuncoro
- Pharmaceutical Research and Development Laboratory of Farmaka Tropis, Faculty of Pharmacy, Universitas Mulawarman, Samarinda, East Kalimantan, 75119, Indonesia.
| | - Deden Indra Dinata
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Bhakti Kencana University, Soekarno-Hatta 754, Bandung, West Java, 40286, Indonesia.
| | - Yoshihito Shiono
- Department of Life, Food, and Environmental Sciences, Faculty of Agriculture, Yamagata University, Wakaba-machi 1-23, Tsuruoka, Yamagata, 997-8555, Japan.
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
5
|
Ren R, Wang X, Leas DA, Scheurer C, Hoevel S, Cal M, Chen G, Zhong L, Katneni K, Pham T, Patil R, Sil D, Walters MJ, Schulze TT, Neville AJ, Dong Y, Wittlin S, Kaiser M, Davis PH, Charman SA, Vennerstrom JL. Antimalarial Dibenzannulated Medium-Ring Keto Lactams. ACS Infect Dis 2023; 9:1964-1980. [PMID: 37695781 PMCID: PMC10860121 DOI: 10.1021/acsinfecdis.3c00245] [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] [Indexed: 09/13/2023]
Abstract
We discovered dibenzannulated medium-ring keto lactams (11,12-dihydro-5H-dibenzo[b,g]azonine-6,13-diones) as a new antimalarial chemotype. Most of these had chromatographic LogD7.4 values ranging from <0 to 3 and good kinetic solubilities (12.5 to >100 μg/mL at pH 6.5). The more polar compounds in the series (LogD7.4 values of <2) had the best metabolic stability (CLint values of <50 μL/min/mg protein in human liver microsomes). Most of the compounds had relatively low cytotoxicity, with IC50 values >30 μM, and there was no correlation between antiplasmodial activity and cytotoxicity. The four most potent compounds had Plasmodium falciparum IC50 values of 4.2 to 9.4 nM and in vitro selectivity indices of 670 to >12,000. They were more than 4 orders-of-magnitude less potent against three other protozoal pathogens (Trypanosoma brucei rhodesiense, Trypanosoma cruzi, and Leishmania donovani) but did have relatively high potency against Toxoplasma gondii, with IC50 values ranging from 80 to 200 nM. These keto lactams are converted into their poorly soluble 4(1H)-quinolone transannular condensation products in vitro in culture medium and in vivo in mouse blood. The similar antiplasmodial potencies of three keto lactam-quinolone pairs suggest that the quinolones likely contribute to the antimalarial activity of the lactams.
Collapse
Affiliation(s)
- Rongguo Ren
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Xiaofang Wang
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Derek A Leas
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Christian Scheurer
- Department of Medical Parasitology and Infection Biology, Swiss Tropical Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Sarah Hoevel
- Department of Medical Parasitology and Infection Biology, Swiss Tropical Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Monica Cal
- Department of Medical Parasitology and Infection Biology, Swiss Tropical Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Longjin Zhong
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Thao Pham
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Rahul Patil
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Diptesh Sil
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Matthias J Walters
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St., Omaha, Nebraska 68182, United States
| | - Thomas T Schulze
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St., Omaha, Nebraska 68182, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Andrew J Neville
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St., Omaha, Nebraska 68182, United States
| | - Yuxiang Dong
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Marcel Kaiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St., Omaha, Nebraska 68182, United States
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jonathan L Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| |
Collapse
|
6
|
Ezzatkhah F, Mahmoudvand H, Raziani Y. The role of Curcuma longa essential oil in controlling acute toxoplasmosis by improving the immune system and reducing inflammation and oxidative stress. Front Cell Infect Microbiol 2023; 13:1161133. [PMID: 37249978 PMCID: PMC10214415 DOI: 10.3389/fcimb.2023.1161133] [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: 02/07/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023] Open
Abstract
Background Chemotherapy with synthetic drugs is the principal approach for toxoplasmosis treatment; however, recent studies reported the limitations and adverse side effects of these chemical drugs. Objective This study aimed to examine the in vitro and in vivo effects of Curcuma longa essential oil (CLE) against the Toxoplasma gondii RH strain. Methods The in vitro effect of different concentrations of CLE on T. gondii tachyzoites was assessed by cell viability assay. Flow cytometry and apoptosis analysis were performed, and nitric oxide production by CLE was also evaluated in tachyzoites. BALB/c mice were orally treated with various doses (1.25, 2.5, and 5 mg·kg-1·day-1) of CLE for 2 weeks. After the induction of acute toxoplasmosis in the mice, their survival rate and the mean number of peritoneal parasites were checked. The hepatic level of antioxidant enzymes and oxidative stress markers was evaluated by commercial kits. The mRNA expression level of proinflammatory cytokines such as interleukin 1-beta (IL-1β) and interferon-gamma (IFN-γ) was evaluated by quantitative real-time PCR. Results CLE, especially at 50 µg/ml, showed potent inhibitory effects on T. gondii tachyzoites. It increased the survival rate (ninth day) and reduced the mean number of peritoneal tachyzoites in the infected mice. CLE dependently increased (p < 0.01) the number of necrotic and apoptotic cells as well as NO production. CLE significantly (p < 0.05) reduced the hepatic level of oxidative stress markers but increased (p < 0.001) the antioxidant enzymes and proinflammatory cytokines in the infected mice, with no important toxicity for vital organs. Conclusion The findings of this survey revealed the significant in vitro inhibitory effects of CLE on T. gondii tachyzoites. The results also exhibited promising in vivo effects of CLE. CLE improved the survival rate of infected mice and reduced the parasite number in them. Although the mechanisms of action of CLE are not clear, our study demonstrated its beneficial effects on acute toxoplasmosis by strengthening the immune system and reducing inflammation and oxidative stress. Still, more studies are required to confirm these results.
Collapse
Affiliation(s)
- Fatemeh Ezzatkhah
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Hossein Mahmoudvand
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Yosra Raziani
- Nursing Department, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| |
Collapse
|
7
|
Anti-Toxoplasma gondii agent isolated from Orostachys malacophylla (Pallas) Fischer. Exp Parasitol 2022; 242:108397. [DOI: 10.1016/j.exppara.2022.108397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/17/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022]
|
8
|
Contopoulos‐Ioannidis DG, Gianniki M, Ai‐Nhi Truong A, Montoya JG. Toxoplasmosis and Schizophrenia: A Systematic Review and Meta‐Analysis of Prevalence and Associations and Future Directions. PSYCHIATRIC RESEARCH AND CLINICAL PRACTICE 2022; 4:48-60. [PMID: 36254187 PMCID: PMC9558922 DOI: 10.1176/appi.prcp.20210041] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Despina G. Contopoulos‐Ioannidis
- Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA (D. G. Contopoulos‐Ioannidis); Department of Pediatrics, University Hospital, Athens, Greece (M. Gianniki); Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA (A. Ai‐Nhi Truong); Dr. Jack S. Remington Laboratory for Specialty Diagnostics, National Reference Center for the Study and Diagnosis of Toxoplasmosis Palo Alto Medical
| | - Maria Gianniki
- Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA (D. G. Contopoulos‐Ioannidis); Department of Pediatrics, University Hospital, Athens, Greece (M. Gianniki); Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA (A. Ai‐Nhi Truong); Dr. Jack S. Remington Laboratory for Specialty Diagnostics, National Reference Center for the Study and Diagnosis of Toxoplasmosis Palo Alto Medical
| | - Angeline Ai‐Nhi Truong
- Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA (D. G. Contopoulos‐Ioannidis); Department of Pediatrics, University Hospital, Athens, Greece (M. Gianniki); Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA (A. Ai‐Nhi Truong); Dr. Jack S. Remington Laboratory for Specialty Diagnostics, National Reference Center for the Study and Diagnosis of Toxoplasmosis Palo Alto Medical
| | - Jose G. Montoya
- Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA (D. G. Contopoulos‐Ioannidis); Department of Pediatrics, University Hospital, Athens, Greece (M. Gianniki); Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, USA (A. Ai‐Nhi Truong); Dr. Jack S. Remington Laboratory for Specialty Diagnostics, National Reference Center for the Study and Diagnosis of Toxoplasmosis Palo Alto Medical
| |
Collapse
|
9
|
A Histone Deacetylase (HDAC) Inhibitor with Pleiotropic In Vitro Anti- Toxoplasma and Anti- Plasmodium Activities Controls Acute and Chronic Toxoplasma Infection in Mice. Int J Mol Sci 2022; 23:ijms23063254. [PMID: 35328672 PMCID: PMC8952293 DOI: 10.3390/ijms23063254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
Toxoplasmosis is a highly prevalent human disease, and virulent strains of this parasite emerge from wild biotopes. Here, we report on the potential of a histone deacetylase (HDAC) inhibitor we previously synthesized, named JF363, to act in vitro against a large panel of Toxoplasma strains, as well as against the liver and blood stages of Plasmodium parasites, the causative agents of malaria. In vivo administration of the drug significantly increases the survival of mice during the acute phase of infection by T. gondii, thus delaying its spreading. We further provide evidence of the compound’s efficiency in controlling the formation of cysts in the brain of T. gondii-infected mice. A convincing docking of the JF363 compound in the active site of the five annotated ME49 T. gondii HDACs was performed by extensive sequence–structure comparison modeling. The resulting complexes show a similar mode of binding in the five paralogous structures and a quite similar prediction of affinities in the micromolar range. Altogether, these results pave the way for further development of this compound to treat acute and chronic toxoplasmosis. It also shows promise for the future development of anti-Plasmodium therapeutic interventions.
Collapse
|
10
|
Hajj RE, Tawk L, Itani S, Hamie M, Ezzeddine J, El Sabban M, El Hajj H. Toxoplasmosis: Current and Emerging Parasite Druggable Targets. Microorganisms 2021; 9:microorganisms9122531. [PMID: 34946133 PMCID: PMC8707595 DOI: 10.3390/microorganisms9122531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Toxoplasmosis is a prevalent disease affecting a wide range of hosts including approximately one-third of the human population. It is caused by the sporozoan parasite Toxoplasma gondii (T. gondii), which instigates a range of symptoms, manifesting as acute and chronic forms and varying from ocular to deleterious congenital or neuro-toxoplasmosis. Toxoplasmosis may cause serious health problems in fetuses, newborns, and immunocompromised patients. Recently, associations between toxoplasmosis and various neuropathies and different types of cancer were documented. In the veterinary sector, toxoplasmosis results in recurring abortions, leading to significant economic losses. Treatment of toxoplasmosis remains intricate and encompasses general antiparasitic and antibacterial drugs. The efficacy of these drugs is hindered by intolerance, side effects, and emergence of parasite resistance. Furthermore, all currently used drugs in the clinic target acute toxoplasmosis, with no or little effect on the chronic form. In this review, we will provide a comprehensive overview on the currently used and emergent drugs and their respective parasitic targets to combat toxoplasmosis. We will also abridge the repurposing of certain drugs, their targets, and highlight future druggable targets to enhance the therapeutic efficacy against toxoplasmosis, hence lessening its burden and potentially alleviating the complications of its associated diseases.
Collapse
Affiliation(s)
- Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, P.O. Box 11-5020, Riad El Solh, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Jana Ezzeddine
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
11
|
Warner RC, Chapman RC, Davis BN, Davis PH. REVIEW OF DNA VACCINE APPROACHES AGAINST THE PARASITE TOXOPLASMA GONDII. J Parasitol 2021; 107:882-903. [PMID: 34852176 DOI: 10.1645/20-157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite that affects both humans and livestock. Transmitted to humans through ingestion, it is the second-leading cause of foodborne illness-related death. Currently, there exists no approved vaccine for humans or most livestock against the parasite. DNA vaccines, a type of subunit vaccine which uses segments of the pathogen's DNA to generate immunity, have shown varying degrees of experimental efficacy against infection caused by the parasite. This review compiles DNA vaccine efforts against Toxoplasma gondii, segmenting the analysis by parasite antigen, as well as a review of concomitant adjuvant usage. No single antigenic group was consistently more effective within in vivo trials relative to others.
Collapse
Affiliation(s)
- Rosalie C Warner
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Ryan C Chapman
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Brianna N Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| |
Collapse
|
12
|
Assessment of the Activity of Decoquinate and Its Quinoline- O-Carbamate Derivatives against Toxoplasma gondii In Vitro and in Pregnant Mice Infected with T. gondii Oocysts. Molecules 2021; 26:molecules26216393. [PMID: 34770802 PMCID: PMC8587999 DOI: 10.3390/molecules26216393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
The quinolone decoquinate (DCQ) is widely used in veterinary practice for the treatment of bacterial and parasitic infections, most notably, coccidiosis in poultry and in ruminants. We have investigated the effects of treatment of Toxoplasma gondii in infected human foreskin fibroblasts (HFF) with DCQ. This induced distinct alterations in the parasite mitochondrion within 24 h, which persisted even after long-term (500 nM, 52 days) treatment, although there was no parasiticidal effect. Based on the low half-maximal effective concentration (IC50) of 1.1 nM and the high selectivity index of >5000, the efficacy of oral treatment of pregnant mice experimentally infected with T. gondii oocysts with DCQ at 10 mg/kg/day for 5 days was assessed. However, the treatment had detrimental effects, induced higher neonatal mortality than T. gondii infection alone, and did not prevent vertical transmission. Thus, three quinoline-O-carbamate derivatives of DCQ, anticipated to have better physicochemical properties than DCQ, were assessed in vitro. One such compound, RMB060, displayed an exceedingly low IC50 of 0.07 nM, when applied concomitantly with the infection of host cells and had no impact on HFF viability at 10 µM. As was the case for DCQ, RMB060 treatment resulted in the alteration of the mitochondrial matrix and loss of cristae, but the changes became apparent at just 6 h after the commencement of treatment. After 48 h, RMB060 induced the expression of the bradyzoite antigen BAG1, but TEM did not reveal any other features reminiscent of bradyzoites. The exposure of infected cultures to 300 nM RMB060 for 52 days did not result in the complete killing of all tachyzoites, although mitochondria remained ultrastructurally damaged and there was a slower proliferation rate. The treatment of mice infected with T. gondii oocysts with RMB060 did reduce parasite burden in non-pregnant mice and dams, but vertical transmission to pups could not be prevented.
Collapse
|
13
|
Virus MA, Ehrhorn EG, Lui LM, Davis PH. Neurological and Neurobehavioral Disorders Associated with Toxoplasma gondii Infection in Humans. J Parasitol Res 2021; 2021:6634807. [PMID: 34712493 PMCID: PMC8548174 DOI: 10.1155/2021/6634807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 09/15/2021] [Indexed: 01/17/2023] Open
Abstract
The intracellular parasite Toxoplasma gondii is estimated to infect up to 30% of the world population, leading to lifelong chronic infection of the brain and muscle tissue. Although most latent T. gondii infections in humans have traditionally been considered asymptomatic, studies in rodents suggest phenotypic neurological changes are possible. Consequently, several studies have examined the link between T. gondii infection and diseases such as schizophrenia, epilepsy, depression, bipolar disorder, dysphoria, Alzheimer's disease, Parkinson's disease, and obsessive-compulsive disorder (OCD). To date, there is varying evidence of the relationship of T. gondii to these human neurological or neurobehavioral disorders. A thorough review of T. gondii literature was conducted to highlight and summarize current findings. We found that schizophrenia was most frequently linked to T. gondii infection, while sleep disruption showed no linkage to T. gondii infection, and other conditions having mixed support for a link to T. gondii. However, infection as a cause of human neurobehavioral disease has yet to be firmly established.
Collapse
Affiliation(s)
- Maxwell A. Virus
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Evie G. Ehrhorn
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - LeeAnna M. Lui
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| |
Collapse
|
14
|
Analogs of marinopyrrole A show enhancement to observed in vitro potency against acute Toxoplasma gondii infection. Antimicrob Agents Chemother 2021; 66:e0079421. [PMID: 34662196 DOI: 10.1128/aac.00794-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The apicomplexan parasite Toxoplasma gondii is the causative agent of toxoplasmosis, a globally distributed infection with severe clinical consequences for immunocompromised individuals and developing fetuses. There are few available treatments, and these are associated with potentially severe adverse effects. Marinopyrrole A, a compound discovered in a marine Streptomyces species, has previously been found to exhibit potent antimicrobial activity, prompting our interest in exploring efficacy against Toxoplasma gondii. We found that marinopyrrole A was a highly potent anti-Toxoplasma molecule, with an in vitro 50% maximal inhibitory concentration (IC50) of 0.31 μM corresponding to a higher potency than that of the current standard of care (pyrimethamine); however, addition of 20% serum led to abrogation of potency, and toxicity to human cell lines was observed. Yet, application of marinopyrrole A to an in vivo lethal acute infection model facilitated significantly enhanced survival at doses of 5, 10, and 20 mg/kg. We then tested a series of marinopyrrole A analogs-RL002, RL003, and RL125-demonstrating significantly increased potency in vitro, with IC50 values ranging from 0.09-0.17 μM (3.6-6.8X increase relative to pyrimethamine). No detectable cytotoxicity was observed up to 50 μM in human foreskin fibroblasts, with cytotoxicity in HepG2 cells ranging from ∼28-50 μM, corresponding to >200X selectivity for parasites over host cells. All analogs additionally showed reduced sensitivity to serum. Further, RL003 potently inhibited in vitro-generated bradyzoites at 0.245 μM. Taken together, these data support further development of marinopyrrole A analogs as promising anti-Toxoplasma molecules to further combat this prevalent infection.
Collapse
|
15
|
Valproic acid inhibits chronic Toxoplasma infection and associated brain inflammation in mice. Antimicrob Agents Chemother 2021; 65:e0100321. [PMID: 34339265 DOI: 10.1128/aac.01003-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Individuals infected with Toxoplasma gondii (T. gondii) are prone to psycho-behavioral disorders, most notably schizophrenia and bipolar. Valproic acid reportedly inhibited the proliferation of T. gondii tachyzoites in vitro. However, animals treated with the drug neither lived longer during acute infection nor had fewer brain cysts upon chronic infection. In this study, a quantitative real-time PCR (qPCR) method was applied to quantify copy numbers of BAG1 (a bradyzoite-specific protein), REP529 DNA (a repetitive DNA fragment of the parasite), and SAG1 (a highly expressed tachyzoite-specific surface protein) in brains of chronically infected mice treated by valproic acid. The treatment inhibited the infection and decreased BAG1, SAG1, and REP529 copy numbers in mice brains (P < 0.0001), comparable to Trimethoprim/Sulfamethoxazole (TMP/SMZ), the common medication for Toxoplasmosis treatment. Moreover, valproic acid decreased brain TNF-α expression (P < 0.0001), comparable to TMP/SMZ. Histological examination of mice brains showed a marked reduction in cyst establishment, perivascular infiltration of lymphocytes, and glial nodules to the same level as the TMP/SMZ group. Our results provide direct evidence for the efficacy of valproic acid, a mood-stabilizing and antipsychotic drug against chronic Toxoplasma infection. These results might help modulate therapeutic regimens for neuropsychiatric patients and design more effective anti-Toxoplasma drugs.
Collapse
|
16
|
Leas DA, Sanford AG, Wu J, Cal M, Kaiser M, Wittlin S, Hemsley RM, Darner EB, Lui LM, Davis PH, Vennerstrom JL. Diaryl Ureas as an Antiprotozoal Chemotype. ACS Infect Dis 2021; 7:1578-1583. [PMID: 33971090 DOI: 10.1021/acsinfecdis.1c00135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We now describe the physicochemical profiling, in vitro ADME, and antiparasitic activity of eight N,N'-diarylureas to assess their potential as a broad-spectrum antiprotozoal chemotype. Chromatographic LogD7.4 values ranged from 2.5 to 4.5; kinetic aq. solubilities were ≤6.3 μg/mL, and plasma protein binding ranged from 95 to 99%. All of the compounds had low intrinsic clearance values in human, but not mouse, liver microsomes. Although no N,N'-diarylurea had submicromolar potency against Trypanosoma cruzi, two had submicromolar potencies against Toxoplasma gondii and Trypanosoma brucei rhodesiense, and five had submicromolar potencies against Leishmania donovani. Plasmodium falciparum appeared to be the most susceptible to growth inhibition by this compound series. Most of the N,N'-diarylureas had antiprotozoal selectivities ≥10. One N,N'-diarylurea had demonstrable activity in mouse models of malaria and toxoplasmosis.
Collapse
Affiliation(s)
- Derek A. Leas
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Austin G. Sanford
- Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States
| | - Jianbo Wu
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| | - Monica Cal
- University of Basel, CH-4003 Basel, Switzerland
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
| | - Marcel Kaiser
- University of Basel, CH-4003 Basel, Switzerland
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
| | - Sergio Wittlin
- University of Basel, CH-4003 Basel, Switzerland
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
| | - Ryan M. Hemsley
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States
| | - Elyssa B. Darner
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States
| | - LeeAnna M. Lui
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska 68182, United States
| | - Jonathan L. Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, 986125 Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| |
Collapse
|
17
|
Păunescu E, Boubaker G, Desiatkina O, Anghel N, Amdouni Y, Hemphill A, Furrer J. The quest of the best - A SAR study of trithiolato-bridged dinuclear Ruthenium(II)-Arene compounds presenting antiparasitic properties. Eur J Med Chem 2021; 222:113610. [PMID: 34144354 DOI: 10.1016/j.ejmech.2021.113610] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/18/2021] [Accepted: 06/01/2021] [Indexed: 12/31/2022]
Abstract
A structure activity relationship (SAR) study of a library of 56 compounds (54 ruthenium and 2 osmium derivatives) based on the trithiolato-bridged dinuclear ruthenium(II)-arene scaffold (general formula [(η6-arene)2Ru2(μ2-SR)3]+, symmetric and [(η6-arene)2Ru2(μ2-SR1)2(μ2-SR2)]+, mixed, respectively) is reported. The 56 compounds (of which 34 are newly designed drug candidates) were synthesized by introducing chemical modifications at the level of bridge thiols, and they were grouped into eight families according to their structural features. The selected fittings were guided by previous results and focused on a fine-tuning of the physico-chemical and steric properties. Newly synthesized complexes were characterized by NMR spectroscopy, mass spectrometry and elemental analysis, and four single-crystal X-ray structures were obtained. The in vitro biological assessment of the compounds was realized by applying a three-step screening cascade: (i) evaluation of the activity against Toxoplasma gondii RH strain tachyzoites expressing β-galactosidase (T. gondii-β-gal) grown in human foreskin fibroblast monolayers (HFF) and assessment of toxicity in non-infected HFF host cells; (ii) dose-response assays using selected compound, and (iii) studies on the effects in murine splenocytes. A primary screening was performed at 1 and 0.1 μM, and resulted in the selection of 39 compounds that inhibited parasite proliferation at 1 μM by more than 95% and reduced the viability of HFF by less than 49%. In the secondary screening, dose-response assays showed that the selected compounds exhibited half maximal inhibitory concentration (IC50) values for T. gondii-β-gal between 0.01 μM and 0.45 μM, with 30 compounds displaying an IC50 lower than 0.1 μM. When applied to non-infected HFF monolayers at 2.5 μM, 8 compounds caused more than 90% and 31 compounds more than 30% viability impairment. The tertiary screening included 14 compounds that did not cause HFF viability loss higher than 50% at 2.5 μM. These derivatives were assessed for potential immunosuppressive activities. First, splenocyte viability was assessed after treatment of cells with concanavalin A (ConA) and lipopolysaccharide (LPS) with compounds applied at 0.1 and 0.5 μM. Subsequently, the 5 compounds exhibiting the lowest splenocyte toxicity were further evaluated for their potential to inhibit B and T cell proliferation. Overall, compound 55 [(η6-p-MeC6H4Pri)2Ru2(μ2-SC6H4-o-CF3)2(μ2-SC6H4-p-OH)]Cl exhibited the most favorable features, and will be investigated as a scaffold for further optimization in terms of anti-parasitic efficacy and drug-like properties.
Collapse
Affiliation(s)
- Emilia Păunescu
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| | - Ghalia Boubaker
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012, Bern, Switzerland
| | - Oksana Desiatkina
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
| | - Nicoleta Anghel
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012, Bern, Switzerland
| | - Yosra Amdouni
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012, Bern, Switzerland; Laboratoire de Parasitologie, Université de la Manouba, Institution de la Recherche et de l'Enseignement Supérieur Agricoles, École Nationale de Médecine Vétérinaire de Sidi Thabet, Sidi Thabet, 2020, Tunisia
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| |
Collapse
|
18
|
Ovung A, Bhattacharyya J. Sulfonamide drugs: structure, antibacterial property, toxicity, and biophysical interactions. Biophys Rev 2021; 13:259-272. [PMID: 33936318 PMCID: PMC8046889 DOI: 10.1007/s12551-021-00795-9] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Sulfonamide (or sulphonamide) functional group chemistry (SN) forms the basis of several groups of drug. In vivo sulfonamides exhibit a range of pharmacological activities, such as anti-carbonic anhydrase and anti-t dihydropteroate synthetase allowing them to play a role in treating a diverse range of disease states such as diuresis, hypoglycemia, thyroiditis, inflammation, and glaucoma. Sulfamethazine (SMZ) is a commonly used sulphonamide drug in veterinary medicine that acts as an antibacterial compound to treat livestock diseases such as gastrointestinal and respiratory tract infections. Sulfadiazine (SDZ) is another frequently employed sulphonamide drug that is used in combination with the anti-malarial drug pyrimethamine to treat toxoplasmosis in warm-blooded animals. This study explores the research findings and the work behaviours of SN (SMZ and SDZ) drugs. The areas covered include SN drug structure, SN drug antibacterial activity, SN drug toxicity, and SN environmental toxicity.
Collapse
Affiliation(s)
- Aben Ovung
- Department of Chemistry, National Institute of Technology Nagaland, Chumukedima, Dimapur, 797103 India
| | - Jhimli Bhattacharyya
- Department of Chemistry, National Institute of Technology Nagaland, Chumukedima, Dimapur, 797103 India
| |
Collapse
|
19
|
In Vitro Selection Implicates ROP1 as a Resistance Gene for an Experimental Therapeutic Benzoquinone Acyl Hydrazone in Toxoplasma gondii. Antimicrob Agents Chemother 2021; 65:AAC.01040-20. [PMID: 33361291 DOI: 10.1128/aac.01040-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/15/2020] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is a globally distributed apicomplexan parasite and the causative agent of toxoplasmosis in humans. While pharmaceuticals exist to combat acute infection, they can produce serious adverse reactions, demonstrating a need for enhanced therapies. KG8 is a benzoquinone acyl hydrazone chemotype identified from a previous chemical screen for which we previously showed in vitro and in vivo efficacy against T. gondii However, the genetic target and mechanism of action of KG8 remain unknown. To investigate potential targets, we generated resistant T. gondii lines by chemical mutagenesis followed by in vitro selection. Whole-genome sequencing of resistant clones revealed a P207S mutation in the gene encoding rhoptry organelle protein 1 (ROP1) in addition to two lesser resistance-conferring mutations in the genes for rhoptry organelle protein 8 (ROP8) and a putative ADP/ATP carrier protein (TGGT1_237700). Expressing ROP1P207S in parental parasites was sufficient to confer significant (10.3-fold increased half-maximal effective concentration [EC50]) KG8 resistance. After generating a library of mutants carrying hypermutated rop1 alleles followed by KG8 pressure, we sequenced the most resistant clonal isolate (>16.9-fold increased EC50) and found independent recapitulation of the P207S mutation, along with three additional mutations in the same region. We also demonstrate that a rop1 knockout strain is insensitive to KG8. These data implicate ROP1 as a putative resistance gene of KG8. This work further identifies a compound that can be used in future studies to better understand ROP1 function and highlights this novel chemotype as a potential scaffold for the development of improved T. gondii therapeutics.
Collapse
|
20
|
Khalifa MM, Martorelli Di Genova B, McAlpine SG, Gallego-Lopez GM, Stevenson DM, Rozema SD, Monaghan NP, Morris JC, Knoll LJ, Golden JE. Dual-Stage Picolinic Acid-Derived Inhibitors of Toxoplasma gondii. ACS Med Chem Lett 2020; 11:2382-2388. [PMID: 33335660 DOI: 10.1021/acsmedchemlett.0c00267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Toxoplasma gondii causes a prevalent human infection for which only the acute stage has an FDA-approved therapy. To find inhibitors of both the acute stage parasites and the persistent cyst stage that causes a chronic infection, we repurposed a compound library containing known inhibitors of parasitic hexokinase, the first step in the glycolysis pathway, along with a larger collection of new structural derivatives. The focused screen of 22 compounds showed a 77% hit rate (>50% multistage inhibition) and revealed a series of aminobenzamide-linked picolinic acids with submicromolar potency against both T. gondii parasite forms. Picolinic acid 23, designed from an antiparasitic benzamidobenzoic acid class with challenging ADME properties, showed 60-fold-enhanced solubility, a moderate LogD7.4, and a 30% improvement in microsomal stability. Furthermore, isotopically labeled glucose tracing revealed that picolinic acid 23 does not function by hexokinase inhibition. Thus, we report a new probe scaffold to interrogate dual-stage inhibition of T. gondii.
Collapse
Affiliation(s)
- Muhammad M. Khalifa
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| | - Bruno Martorelli Di Genova
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Sarah G. McAlpine
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - Gina M. Gallego-Lopez
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
- Morgridge Institute for Research, Madison, Wisconsin 53715, United States
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States of America
| | - Soren D. Rozema
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| | - Neil P. Monaghan
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - James C. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - Laura J. Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Jennifer E. Golden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| |
Collapse
|
21
|
Marcu IC, Eberhard N, Yerly A, Balmer V, Hemphill A, Mogel H, Gaschen V, Stoffel MH, Bluteau J. Isolation of Human Small Extracellular Vesicles and Tracking of their Uptake by Retinal Pigment Epithelial Cells In Vitro. Int J Mol Sci 2020; 21:E3799. [PMID: 32471212 PMCID: PMC7313035 DOI: 10.3390/ijms21113799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022] Open
Abstract
Small extracellular vesicles (EVs) are among the most frequently investigated EVs and play major roles in intercellular communication by delivering various cargo molecules to target cells. They could potentially represent an alternative delivery strategy to treat ocular toxoplasmosis, a parasitosis affecting the retinal pigment epithelium (RPE). To date, the uptake of human small EVs by RPE cells has never been reported. In this study, we report on the intracellular uptake of fluorescently labelled human urine and fibroblast-derived small EVs by human RPE cells. In summary, both dye-labelled urinary small EVs and small EVs obtained from fibroblasts stably expressing membrane-bound green fluorescent protein were successfully internalized by RPE cells as revealed by immunohistochemistry. In recipient ARPE19 cells, BODIPY-labelled small EVs were found in close vicinity to the parasite Toxoplasma gondii. Additionally, an ultrastructural method was enabled to distinguish between labelled exogenous and endogenous small EVs within target cells.
Collapse
Affiliation(s)
- Irene C. Marcu
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| | - Naja Eberhard
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (N.E.); (V.B.); (A.H.)
| | - Anaïs Yerly
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| | - Verena Balmer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (N.E.); (V.B.); (A.H.)
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (N.E.); (V.B.); (A.H.)
| | - Helga Mogel
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| | - Véronique Gaschen
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| | - Michael H. Stoffel
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| | - Jasmin Bluteau
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland; (I.C.M.); (A.Y.); (H.M.); (V.G.); (M.H.S.)
| |
Collapse
|
22
|
Salin NH, Noordin R, Al-Najjar BO, Kamarulzaman EE, Yunus MH, Karim IZA, Nasim NNM, Zakaria II, Wahab HA. Identification of potential dual -targets anti- toxoplasma gondii compounds through structure-based virtual screening and in-vitro studies. PLoS One 2020; 15:e0225232. [PMID: 32442170 PMCID: PMC7244133 DOI: 10.1371/journal.pone.0225232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/23/2020] [Indexed: 11/18/2022] Open
Abstract
Toxoplasma gondii is the etiologic agent of toxoplasmosis, a disease which can lead to morbidity and mortality of the fetus and immunocompromised individuals. Due to the limited effectiveness or side effects of existing drugs, the search for better drug candidates is still ongoing. In this study, we performed structure-based screening of potential dual-targets inhibitors of active sites of T. gondii drug targets such as uracil phosphoribosyltransferase (UPRTase) and adenosine kinase (AK). First screening of virtual compounds from the National Cancer Institute (NCI) was performed via molecular docking. Subsequently, the hit compounds were tested in-vitro for anti- T. gondii effect using cell viability assay with Vero cells as host to determine cytotoxicity effects and drug selectivities. Clindamycin, as positive control, showed a selectivity index (SI) of 10.9, thus compounds with SI > 10.9 specifically target T. gondii proliferation with no significant effect on the host cells. Good anti- T. gondii effects were observed with NSC77468 (7-ethoxy-4-methyl-6,7-dihydro-5H-thiopyrano[2,3-d]pyrimidin-2-amine) which showed SI values of 25. This study showed that in-silico selection can serve as an effective way to discover potentially potent and selective compounds against T. gondii.
Collapse
Affiliation(s)
- Nurul Hanim Salin
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Gelugor, Pulau Pinang, Malaysia
| | - Rahmah Noordin
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
| | - Belal O. Al-Najjar
- Faculty of Pharmacy Al-Ahliyya Amman, University Amman Jordan, Amman, Jordan
| | | | - Muhammad Hafiznur Yunus
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
| | | | - Nurul Nadieya Mohd Nasim
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Gelugor, Pulau Pinang, Malaysia
| | - Iffah Izzati Zakaria
- Synthetic Biology & Cell Factories, Malaysia Genome Institute, National Institutes of Biotechnology Malaysia, Selangor, Gelugor, Malaysia
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
- * E-mail:
| |
Collapse
|
23
|
Njiri OA, Zhang X, Zhang Y, Wu B, Jiang L, Li Q, Liu W, Chen T. CD209 C-Type Lectins Promote Host Invasion, Dissemination, and Infection of Toxoplasma gondii. Front Immunol 2020; 11:656. [PMID: 32391004 PMCID: PMC7190871 DOI: 10.3389/fimmu.2020.00656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/23/2020] [Indexed: 01/24/2023] Open
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis and a major opportunistic parasite associated with AIDS, is able to invade host cells of animals and humans. Studies suggested that the ability of host invasion by the tachyzoite, the infectious form of T. gondii, is essential for the pathogenicity to promote its dissemination to other parts of animal hosts. However, the detailed molecular mechanisms for host invasion and dissemination of the parasites are not clear. On the other hand, viruses and bacteria are able to interact with and hijack DC-SIGN (CD209) C-type lectin on antigen presenting cells (APCs), such as dendritic cells and macrophages as the Trojan horses to promote host dissemination. In this study, we showed that invasion of T. gondii into host cells was enhanced by this parasite-CD209 interaction that were inhibited by ligand mimicking-oligosaccharides and the anti-CD209 antibody. Furthermore, covering the exposures of DC-SIGN by these oligosaccharides reduced parasite burden, host spreading and mortality associated with T. gondii infection. These results suggested that interaction of T. gondii to APCs expressing DC-SIGN might promote host dissemination and infection. Can the blockage of this interaction with Mannan and/or anti-CD209 antibody be developed as a prevention or treatment method for T. gondii infection?
Collapse
Affiliation(s)
- Olivia Adhiambo Njiri
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.,Department of Biological Sciences, Faculty of Science, Engineering and Technology, Chuka University, Chuka, Kenya
| | - Xiaoyan Zhang
- Division of Parasitology, Department of Pathogen Biology, School of Basic Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingmiao Zhang
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Bicong Wu
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Lingyu Jiang
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qiao Li
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Wenqi Liu
- Division of Parasitology, Department of Pathogen Biology, School of Basic Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tie Chen
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| |
Collapse
|
24
|
Abugri DA, Witola WH. Interaction of apigenin-7-O-glucoside with pyrimethamine against Toxoplasma gondii growth. J Parasit Dis 2020; 44:221-229. [PMID: 32174728 PMCID: PMC7046874 DOI: 10.1007/s12639-019-01185-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 11/25/2019] [Indexed: 12/24/2022] Open
Abstract
Apigenin-7-O-glucoside, a flavonoid glucoside known to inhibit cancer cell growth, fungi growth, both intra and extracellular reactive oxygen species generation, causing cell arrest and damage to the plasma membrane, was tested alone or in combination with a dihydrofolate inhibitor (pyrimethamine) against Toxoplasma gondii (T. gondii) growth. The anti-T. gondii activity was carried out using a high throughput antiparasitic drug screening cell-based assay known as 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H tetrazolium, monosodium salt (WST-8) and fluorescence plate reader. The 50% effective concentration inhibition and 95% confidence interval values for individual and combination treatments against T. gondii were 0.80 (0.38-1.29) µg/mL, 1.05 (0.275-2.029) µg/mL, and 0.40 (0-1.06) µg/mL for apigenin-7-O-glucoside, pyrimethamine, and apigenin-7-O-glucoside plus pyrimethamine, respectively. Interestingly, the apigenin-7-O-glucoside plus pyrimethamine combination showed an additive inhibition effect against T. gondii growth in vitro using the fractional inhibitory concentration index method. It was discovered that the apigenin-7-O-glucoside combination with pyrimethamine had a high selectivity index 62.5, which implies 62-fold inhibition activity against the parasite versus human foreskin fibroblast cell cytotoxicity. This new combination hit is novel and will have the potential for future effective, safe, and less costly anti-Toxoplasma drug development, if its in vivo activity shows similar findings.
Collapse
Affiliation(s)
- Daniel A. Abugri
- Department of Chemistry, Laboratory of Ethnomedicine, Parasitology and Drug Discovery, Tuskegee University, Tuskegee, AL 36088 USA
- Department of Biology, Tuskegee University, Tuskegee, AL 36088 USA
| | - William H. Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, 2432 VMBSB, MC-002, 2001 South Lincoln Avenue, Urbana, Champaign, IL USA
| |
Collapse
|
25
|
Lapinskas PJ, Ben-Harari RR. Perspective on current and emerging drugs in the treatment of acute and chronic toxoplasmosis. Postgrad Med 2019; 131:589-596. [PMID: 31399001 DOI: 10.1080/00325481.2019.1655258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
No new drugs for treatment of toxoplasmosis have been approved in over 60 years, despite the burden of toxoplasmosis on human society. The small selection of effective drugs is limited by important side effects, often limiting patient use. This perspective highlights promising late-stage drug candidates in the treatment of toxoplasmosis. Presently, drugs target the tachyzoite form of the parasite Toxoplasma gondii responsible for the acute infection but do not eradicate the tissue cyst form underlying chronic infection. Pyrimethamine - the first-line and only approved drug for treatment of toxoplasmosis in the United States - inhibits parasite DNA synthesis by inhibiting dihydrofolate reductase (DHFR). Two novel DHFR inhibitors with improved potency and selectivity for parasite DHFR over human DHFR are in clinical-stage development. One of the most advanced and promising therapeutic targets, demonstrating potential to treat both acute and chronic toxoplasmosis, is the calcium-dependent protein kinase 1 (CDPK1) which plays an essential role in the intracellular replicative cycle of the parasite, and has no direct mammalian homolog. Two CDPK1 inhibitor programs have identified potent and selective lead series, demonstrating acceptable systemic and CNS exposure, and in vivo efficacy in animal models of acute and chronic infection. Physicians need a better arsenal of parasiticidal drugs for the treatment of toxoplasmosis, particularly those active against tissue cysts.
Collapse
|
26
|
Konstantinovic N, Guegan H, Stäjner T, Belaz S, Robert-Gangneux F. Treatment of toxoplasmosis: Current options and future perspectives. Food Waterborne Parasitol 2019; 15:e00036. [PMID: 32095610 PMCID: PMC7033996 DOI: 10.1016/j.fawpar.2019.e00036] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 02/08/2023] Open
Abstract
Toxoplasmosis is a worldwide parasitic disease infecting about one third of humans, with possible severe outcomes in neonates and immunocompromised patients. Despite continuous and successful efforts to improve diagnosis, therapeutic schemes have barely evolved since many years. This article aims at reviewing the main clinical trials and current treatment practices, and at addressing future perspectives in the light of ongoing researches.
Collapse
Affiliation(s)
- Neda Konstantinovic
- National Reference Laboratory for Toxoplasmosis, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia
| | - Hélène Guegan
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000 Rennes, France
| | - Tijana Stäjner
- National Reference Laboratory for Toxoplasmosis, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia
| | - Sorya Belaz
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000 Rennes, France
| | | |
Collapse
|
27
|
Derivatives of a benzoquinone acyl hydrazone with activity against Toxoplasma gondii. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2018; 8:488-492. [PMID: 30500526 PMCID: PMC6262783 DOI: 10.1016/j.ijpddr.2018.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 12/22/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite with global incidence. The acute infection, toxoplasmosis, is treatable but current regimens have poor host tolerance and no cure has been found for latent infections. This work builds upon a previous high throughput screen which identified benzoquinone acyl hydrazone (KG8) as the most promising compound; KG8 displayed potent in vitro activity against T. gondii but only marginal in vivo efficacy in a T. gondii animal model. To define the potential of this new lead compound, we now describe a baseline structure-activity relationship for this chemotype. Several derivatives displayed IC50's comparable to that of the control treatment pyrimethamine with little to no cytotoxicity. The best of these, KGW44 and KGW59, had higher metabolic stability than KG8. In an in vivo T. gondii murine model, KGW59 significantly increased survivorship. This work provides new insights for optimization of this novel chemotype. Derivatives from a previously described anti T. gondii compound were characterized. Top derivatives showed increased activity against T. gondii in vitro. Derivatives demonstrated low cytotoxicity in host cell lines. Derivative KGW59 showed efficacy in vivo with a strong therapeutic window. These early-leads are suitable for further development as toxoplasmosis therapies.
Collapse
|
28
|
Rocha-Roa C, Molina D, Cardona N. A Perspective on Thiazolidinone Scaffold Development as a New Therapeutic Strategy for Toxoplasmosis. Front Cell Infect Microbiol 2018; 8:360. [PMID: 30386743 PMCID: PMC6198644 DOI: 10.3389/fcimb.2018.00360] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022] Open
Abstract
Toxoplasma gondii is one of the most successful parasites due to its ability to infect a wide variety of warm-blooded animals. It is estimated that one-third of the world's population is latently infected. The generic therapy for toxoplasmosis has been a combination of antifolates such as pyrimethamine or trimethoprim with either sulfadiazine or antibiotics such as clindamycin with a combination with leucovorin to prevent hematologic toxicity. This therapy shows limitations such as drug intolerance, low bioavailability or drug resistance by the parasite. There is a need for the development of new molecules with the capacity to block any stage of the parasite's life cycle in humans or in a different type of hosts. Heterocyclic compounds are promissory drugs due to its reported biological activity; for this reason, thiazolidinone and its derivatives are presented as a new alternative not only for its inhibitory activity against the parasite but also for its high selectivity-level with high therapeutic index. Thiazolidinones are an important scaffold known to be associated with anticancer, antibacterial, antifungal, antiviral, antioxidant, and antidiabetic activities. The molecule possesses an imidazole ring that has been described as an antiprotozoal agent with antiparasitic properties and less toxicity. Thiazolidinone derivatives have been reportedly as building blocks in organic chemistry and as scaffolds for drug discovery. Here we present a perspective of how structural modifications of the thiazolidinone core could generate new compounds with high anti-parasitic effect and less toxic results.
Collapse
Affiliation(s)
- Cristian Rocha-Roa
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia
| | - Diego Molina
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia
| | - Néstor Cardona
- Centre for Biomedical Research CIBM, University of Quindío, Armenia, Colombia.,Dentistry Faculty, University Antonio Nariño, Armenia, Colombia
| |
Collapse
|
29
|
Treatment of Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Clin Microbiol Rev 2018; 31:31/4/e00057-17. [PMID: 30209035 DOI: 10.1128/cmr.00057-17] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Primary Toxoplasma gondii infection is usually subclinical, but cervical lymphadenopathy or ocular disease can be present in some patients. Active infection is characterized by tachyzoites, while tissue cysts characterize latent disease. Infection in the fetus and in immunocompromised patients can cause devastating disease. The combination of pyrimethamine and sulfadiazine (pyr-sulf), targeting the active stage of the infection, is the current gold standard for treating toxoplasmosis, but failure rates remain significant. Although other regimens are available, including pyrimethamine in combination with clindamycin, atovaquone, clarithromycin, or azithromycin or monotherapy with trimethoprim-sulfamethoxazole (TMP-SMX) or atovaquone, none have been found to be superior to pyr-sulf, and no regimen is active against the latent stage of the infection. Furthermore, the efficacy of these regimens against ocular disease remains uncertain. In multiple studies, systematic screening for Toxoplasma infection during gestation, followed by treatment with spiramycin for acute maternal infections and with pyr-sulf for those with established fetal infection, has been shown to be effective at preventing vertical transmission and minimizing the severity of congenital toxoplasmosis (CT). Despite significant progress in treating human disease, there is a strong impetus to develop novel therapeutics for both the acute and latent forms of the infection. Here we present an overview of toxoplasmosis treatment in humans and in animal models. Additional research is needed to identify novel drugs by use of innovative high-throughput screening technologies and to improve experimental models to reflect human disease. Such advances will pave the way for lead candidates to be tested in thoroughly designed clinical trials in defined patient populations.
Collapse
|
30
|
Chan WC, Wai Chan DH, Lee KW, Tin WS, Wong HN, Haynes RK. Evaluation and optimization of synthetic routes from dihydroartemisinin to the alkylamino-artemisinins artemiside and artemisone: A test of N-glycosylation methodologies on a lipophilic peroxide. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.04.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Sanford AG, Schulze TT, Potluri LP, Hemsley RM, Larson JJ, Judge AK, Zach SJ, Wang X, Charman SA, Vennerstrom JL, Davis PH. Novel Toxoplasma gondii inhibitor chemotypes. Parasitol Int 2018; 67:107-111. [PMID: 29081387 DOI: 10.1016/j.parint.2017.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 11/18/2022]
Abstract
We profiled three novel T. gondii inhibitors identified from an antimalarial phenotypic high throughput screen (HTS) campaign: styryl 4-oxo-1,3-benzoxazin-4-one KG3, tetrahydrobenzo[b]pyran KG7, and benzoquinone hydrazone KG8. These compounds inhibit T. gondii in vitro with IC50 values ranging from 0.3 to 2μM, comparable to that of 0.25 to 1.5μM for the control drug pyrimethamine. KG3 had no measurable cytotoxicity against five mammalian cell lines, whereas KG7 and KG8 inhibited the growth of 2 of 5 cell lines with KG8 being the least selective for T. gondii. None of the compounds were mutagenic in an Ames assay. Experimental gLogD7.4 and calculated PSA values for the three compounds were well within the ranges predicted to be favorable for good ADME, even though each compound had relatively low aqueous solubility. All three compounds were metabolically unstable, especially KG3 and KG7. Multiple IP doses of 5mg/kg KG7 and KG8 increased survival in a T. gondii mouse model. Despite their liabilities, we suggest that these compounds are useful starting points for chemical prospecting, scaffold-hopping, and optimization.
Collapse
Affiliation(s)
- A G Sanford
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - T T Schulze
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - L P Potluri
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - R M Hemsley
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - J J Larson
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - A K Judge
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - S J Zach
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - X Wang
- College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - S A Charman
- Centre for Drug Candidate Optimization, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - J L Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - P H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA.
| |
Collapse
|
32
|
Resistance towards monensin is proposed to be acquired in a Toxoplasma gondii model by reduced invasion and egress activities, in addition to increased intracellular replication. Parasitology 2017; 145:313-325. [PMID: 28870270 DOI: 10.1017/s0031182017001512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Monensin (Mon) is an anticoccidial polyether ionophore widely used to control coccidiosis. The extensive use of polyether ionophores on poultry farms resulted in widespread resistance, but the underlying resistance mechanisms are unknown in detail. For analysing the mode of action by which resistance against polyether ionophores is obtained, we induced in vitro Mon resistance in Toxoplasma gondii-RH strain (MonR-RH) and compared it with the sensitive parental strain (Sen-RH). The proteome assessment of MonR-RH and Sen-RH strains was obtained after isotopic labelling using stable isotope labelling by amino acid in cell culture. Relative proteomic quantification between resistant and sensitive strains was performed using liquid chromatography-mass spectrometry/mass spectrometry. Overall, 1024 proteins were quantified and 52 proteins of them were regulated. The bioinformatic analysis revealed regulation of cytoskeletal and transmembrane proteins being involved in transport mechanisms, metal ion-binding and invasion. During invasion, actin and microneme protein 8 (MIC8) are seem to be important for conoid extrusion and forming moving junction with host cells, respectively. Actin was significantly upregulated, while MIC8 was downregulated, which indicate an invasion reduction in the resistant strain. Resistance against Mon is not a simple process but it involves reduced invasion and egress activity of T. gondii tachyzoites while intracellular replication is enhanced.
Collapse
|
33
|
Montazeri M, Sharif M, Sarvi S, Mehrzadi S, Ahmadpour E, Daryani A. A Systematic Review of In vitro and In vivo Activities of Anti -Toxoplasma Drugs and Compounds (2006-2016). Front Microbiol 2017; 8:25. [PMID: 28163699 PMCID: PMC5247447 DOI: 10.3389/fmicb.2017.00025] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/05/2017] [Indexed: 11/23/2022] Open
Abstract
The currently available anti-Toxoplasma agents have serious limitations. This systematic review was performed to evaluate drugs and new compounds used for the treatment of toxoplasmosis. Data was systematically collected from published papers on the efficacy of drugs/compounds used against Toxoplasma gondii (T. gondii) globally during 2006-2016. The searched databases were PubMed, Google Scholar, Science Direct, ISI Web of Science, EBSCO, and Scopus. One hundred and eighteen papers were eligible for inclusion in this systematic review, which were both in vitro and in vivo studies. Within this review, 80 clinically available drugs and a large number of new compounds with more than 39 mechanisms of action were evaluated. Interestingly, many of the drugs/compounds evaluated against T. gondii act on the apicoplast. Therefore, the apicoplast represents as a potential drug target for new chemotherapy. Based on the current findings, 49 drugs/compounds demonstrated in vitro half-maximal inhibitory concentration (IC50) values of below 1 μM, but most of them were not evaluated further for in vivo effectiveness. However, the derivatives of the ciprofloxacin, endochin-like quinolones and 1-[4-(4-nitrophenoxy) phenyl] propane-1-one (NPPP) were significantly active against T. gondii tachyzoites both in vitro and in vivo. Thus, these compounds are promising candidates for future studies. Also, compound 32 (T. gondii calcium-dependent protein kinase 1 inhibitor), endochin-like quinolones, miltefosine, rolipram abolish, and guanabenz can be repurposed into an effective anti-parasitic with a unique ability to reduce brain tissue cysts (88.7, 88, 78, 74, and 69%, respectively). Additionally, no promising drugs are available for congenital toxoplasmosis. In conclusion, as current chemotherapy against toxoplasmosis is still not satisfactory, development of well-tolerated and safe specific immunoprophylaxis in relaxing the need of dependence on chemotherapeutics is a highly valuable goal for global disease control. However, with the increasing number of high-risk individuals, and absence of a proper vaccine, continued efforts are necessary for the development of novel treatment options against T. gondii. Some of the novel compounds reviewed here may represent good starting points for the discovery of effective new drugs. In further, bioinformatic and in silico studies are needed in order to identify new potential toxoplasmicidal drugs.
Collapse
Affiliation(s)
- Mahbobeh Montazeri
- Toxoplasmosis Research Center, Mazandaran University of Medical SciencesSari, Iran
- Student Research Committee, Mazandaran University of Medical SciencesSari, Iran
| | - Mehdi Sharif
- Toxoplasmosis Research Center, Mazandaran University of Medical SciencesSari, Iran
- Department of Parasitology and Mycology, Sari Medical School, Mazandaran University of Medical SciencesSari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical SciencesSari, Iran
- Department of Parasitology and Mycology, Sari Medical School, Mazandaran University of Medical SciencesSari, Iran
| | - Saeed Mehrzadi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences TehranIran
| | - Ehsan Ahmadpour
- Drug Applied Research Center, Tabriz University of Medical SciencesTabriz, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical SciencesSari, Iran
- Department of Parasitology and Mycology, Sari Medical School, Mazandaran University of Medical SciencesSari, Iran
| |
Collapse
|