1
|
Thiam F, Djoumoi D, Mbaye MN, Fall A, Diouara AAM, Diop M, Nguer CM, Mbengue B, Diop G, Kohli E, Dieye A. Secreted extracellular heat shock protein gp96 and inflammatory cytokines are markers of severe malaria outcome. Cell Stress Chaperones 2024; 30:48-56. [PMID: 39732362 PMCID: PMC11761890 DOI: 10.1016/j.cstres.2024.12.004] [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: 06/03/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
Malaria caused by Plasmodium spp., is a major public health issue in sub-Saharan Africa. The fight against malaria has stalled due to increasing resistance to treatments and insecticides. There is an urgent need to focus on new therapeutic targets to combat malaria effectively. This study aimed to measure the secreted heat shock protein gp96 levels in both malaria patients and controls. Indeed, gp96 plays a crucial role in parasite survival within the host and in establishing a successful infection. Therefore, gp96 could be a promising target for antimalarial drugs. In our study, we included 60 malaria patients, 30 with severe malaria (SM) and 30 with uncomplicated malaria (UM). Additionally, 28 controls were included. Using the ELISA method, we measured gp96 levels in the participants' blood samples. We then used the Mann-Whitney or analyse of variance tests to calculate descriptive statistics and determined the correlation between gp96 level and parasitemia using Spearman's rank correlation test. The study found that gp96 levels in the plasma significantly increased in malaria patients (23.86 ng/mL) compared to control (5.88 ng/mL), with a P < 0.0001. Interestingly, there was a significant difference between SM (27.56 ng/mL) and UM (13.9 ng/mL), with a P-value of 0.001. These findings are accompanied by significantly higher parasitemia and elevated proinflammatory cytokines such as IL-17A and IL-1β levels in SM patients compared to UM and controls. Furthermore, there was no significant positive correlation between gp96 levels and parasitemia/proinflammatory cytokines. Our research has revealed, for the first time, that individuals with SM have significantly higher levels of gp96 in the context of high parasitemia and proinflammatory cytokines. Our preliminary results will be taken further to evaluate gp96 as a valuable biomarker for the diagnosis of SM and a potential target for antimalarial drug discovery.
Collapse
Affiliation(s)
- Fatou Thiam
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal.
| | - Djibaba Djoumoi
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Mame Ndew Mbaye
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Aminata Fall
- Unite postulante de Biologie Genetique, Genomique et Bio-informatique (G2B), Departement de Biologie animale, Faculté des Sciences et Techniques, Universite Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Abou Abdallah Malick Diouara
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Mamadou Diop
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Cheikh Momar Nguer
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Babacar Mbengue
- Service d'Immunologie, Faculté de Médecine, de Pharmacie et d'Odontostomatologie, Université Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Gora Diop
- Unite postulante de Biologie Genetique, Genomique et Bio-informatique (G2B), Departement de Biologie animale, Faculté des Sciences et Techniques, Universite Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Evelyne Kohli
- Université de Bourgogne Franche-Comté, LNC UMR 866 Dijon, France
| | - Alioune Dieye
- Service d'Immunologie, Faculté de Médecine, de Pharmacie et d'Odontostomatologie, Université Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| |
Collapse
|
2
|
Muzenda FL, Stofberg ML, Mthembu W, Achilonu I, Strauss E, Zininga T. Characterization and Inhibition of the Chaperone Function of Plasmodium falciparum Glucose-Regulated Protein 94 kDa (PfGrp94). Proteins 2024. [PMID: 39670568 DOI: 10.1002/prot.26779] [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: 08/22/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Plasmodium falciparum expresses four heat shock protein 90 (Hsp90) members. Among these, one, glucose-regulated protein 94 (PfGrp94), is localized in the endoplasmic reticulum (ER). Both the cytosolic and ER-based Hsp90s are essential for parasite survival under all growth conditions. The cytosolic version has been extensively studied and has been targeted in several efforts through the repurposing of anticancer therapeutics as antimalarial drugs. However, PfGrp94 has not been fully characterized and some of its functions related to the ER stress response are not fully understood. Structural analysis of the recombinant full-length PfGrp94 protein showed a predominantly α-helical secondary structure and its thermal resilience was modulated by 5'-N-ethyl-carboxamide-adenosine (NECA) and nucleotides ATP/ADP. PfGrp94 exhibits ATPase activity and suppressed heat-induced aggregation of a model substrate, malate dehydrogenase, in a nucleotide-dependent manner. However, these PfGrp94 chaperone functions were abrogated by NECA. Molecular docking and molecular dynamics (MD) simulations showed that NECA interacted with unique residues on PfGrp94, which could be potentially exploited for selective drug design. Finally, using parasites maintained at the red blood stage, NECA exhibited moderate antiplasmodial activity (IC50 of 4.3, 7.4, and 10.0 μM) against three different P. falciparum strains. Findings from this study provide the first direct evidence for the correlation between in silico, biochemical, and in vitro data toward utilizing the ER-based chaperone, PfGrp94, as a drug target against the malaria parasites.
Collapse
Affiliation(s)
| | | | - Wendy Mthembu
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Ikechukwu Achilonu
- Protein Structure Function Research Group, University of Witwatersrand, Johannesburg, South Africa
| | - Erick Strauss
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
3
|
Mitra P, Deshmukh AS. Proteostasis is a key driver of the pathogenesis in Apicomplexa. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119824. [PMID: 39168412 DOI: 10.1016/j.bbamcr.2024.119824] [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: 01/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Proteostasis, including protein folding mediated by molecular chaperones, protein degradation, and stress response pathways in organelles like ER (unfolded protein response: UPR), are responsible for cellular protein quality control. This is essential for cell survival as it regulates and reprograms cellular processes. Here, we underscore the role of the proteostasis pathway in Apicomplexan parasites with respect to their well-characterized roles as well as potential roles in many parasite functions, including survival, multiplication, persistence, and emerging drug resistance. In addition to the diverse physiological importance of proteostasis in Apicomplexa, we assess the potential of the pathway's components as chemotherapeutic targets.
Collapse
Affiliation(s)
- Pallabi Mitra
- BRIC-Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| | | |
Collapse
|
4
|
Matlhodi T, Makatsela LP, Dongola TH, Simelane MBC, Shonhai A, Gumede NJ, Mokoena F. Auto QSAR-based active learning docking for hit identification of potential inhibitors of Plasmodium falciparum Hsp90 as antimalarial agents. PLoS One 2024; 19:e0308969. [PMID: 39585817 PMCID: PMC11588265 DOI: 10.1371/journal.pone.0308969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/02/2024] [Indexed: 11/27/2024] Open
Abstract
Malaria which is mainly caused by Plasmodium falciparum parasite remains a devastating public health concern, necessitating the need to develop new antimalarial agents. P. falciparum heat shock protein 90 (Hsp90), is indispensable for parasite survival and a promising drug target. Inhibitors targeting the ATP-binding pocket of the N-terminal domain have anti-Plasmodium effects. We proposed a de novo active learning (AL) driven method in tandem with docking to predict inhibitors with unique scaffolds and preferential selectivity towards PfHsp90. Reference compounds, predicted to bind PfHsp90 at the ATP-binding pocket and possessing anti-Plasmodium activities, were used to generate 10,000 unique derivatives and to build the Auto-quantitative structures activity relationships (QSAR) models. Glide docking was performed to predict the docking scores of the derivatives and > 15,000 compounds obtained from the ChEMBL database. Re-iterative training and testing of the models was performed until the optimum Kennel-based Partial Least Square (KPLS) regression model with a regression coefficient R2 = 0.75 for the training set and squared correlation prediction Q2 = 0.62 for the test set reached convergence. Rescoring using induced fit docking and molecular dynamics simulations enabled us to prioritize 15 ATP/ADP-like design ideas for purchase. The compounds exerted moderate activity towards P. falciparum NF54 strain with IC50 values of ≤ 6μM and displayed moderate to weak affinity towards PfHsp90 (KD range: 13.5-19.9μM) comparable to the reported affinity of ADP. The most potent compound was FTN-T5 (PfN54 IC50:1.44μM; HepG2/CHO cells SI≥ 29) which bound to PfHsp90 with moderate affinity (KD:7.7μM), providing a starting point for optimization efforts. Our work demonstrates the great utility of AL for the rapid identification of novel molecules for drug discovery (i.e., hit identification). The potency of FTN-T5 will be critical for designing species-selective inhibitors towards developing more efficient agents against malaria.
Collapse
Affiliation(s)
- Thato Matlhodi
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| | - Lisema Patrick Makatsela
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| | | | | | - Addmore Shonhai
- Department of Biochemistry & Microbiology, University of Venda, Thohoyandou, South Africa
| | - Njabulo Joyfull Gumede
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University (WSU), Umthatha, Eastern Cape, South Africa
| | - Fortunate Mokoena
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| |
Collapse
|
5
|
Pumpitakkul V, Roytrakul S, Phaonakrop N, Thongphakdee A, Sanannu S, Nipanunt T, Pandhumas S, Kaewsen K, Ploypetch S, Sirisawadi S, Kunnasut N, Anuracpreeda P, Watthanadirek-Wijidwong A, Suriyaphol G. Analysis of serum proteomic profiles of endangered Siamese and Burmese Eld's deer infected with subclinical Babesia bovis in Thailand. Acta Trop 2024; 257:107294. [PMID: 38909725 DOI: 10.1016/j.actatropica.2024.107294] [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: 03/27/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The endangered Eld's deer is a conserved species in Thailand, where tropical parasitic infections are endemic. Although Eld's deer with babesiosis are generally asymptomatic, they can still harbor the parasite and serve as reservoirs for ticks, spreading the infection to healthy animals within the herd. The present study aimed to investigate potential serum proteome biomarkers of Eld's deer with subclinical Babesia bovis infection. A total of 67 blood samples were collected from captive Siamese and Burmese Eld's deer showing no signs of parasitic infection. The nested polymerase chain reaction (nPCR) of a conserved spherical body protein 2 (sbp-2) gene of B. bovis was utilized to classify Eld's deer groups, with 25.37 % (17/67) testing positive for B. bovis. Additionally, the application of proteomic studies showed that six B. bovis proteins, such as Obg-like ATPase 1 (OLA1) and heat shock protein 90 (HSP90), were significantly upregulated by more than a two-fold change compared with the PCR-negative samples. Of the 55 overexpressed serum proteins in the PCR-positives, alpha 2-HS glycoprotein (AHSG) and immunoglobulin lambda variable 2-8 (IGLV2-8) were notably among the top 10 proteins with the highest area under curve (AUC) values. Hence, they were proposed as potential biomarkers for subclinical B. bovis infection in Eld's deer. Analysis of the protein interaction network revealed interactions between Eld's deer AHSG and B. bovis OLA1 and HSP90, alongside associations with other proteins such as erb-b2 receptor tyrosine kinase 2 (ERBB2) and epidermal growth factor receptor (EGFR). These interactions were involved in the immune system pathway and inflammatory responses. Our findings shed light on subclinical infection of B. bovis in Eld's deer and identify potential biomarkers, contributing to the further effective detection and monitoring of B. bovis infection in this endangered species.
Collapse
Affiliation(s)
- Vichayanee Pumpitakkul
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Ampika Thongphakdee
- Animal Conservation and Research Institute, Zoological Park Organization of Thailand under the Royal Patronage of H.M. The King, Bangkok 10800, Thailand
| | - Saowaphang Sanannu
- Animal Conservation and Research Institute, Zoological Park Organization of Thailand under the Royal Patronage of H.M. The King, Bangkok 10800, Thailand
| | - Tarasak Nipanunt
- Huai Kha Khaeng Wildlife Breeding Center, Department of National Parks, Wildlife and Plant Conservation, Uthai Thani 61160, Thailand
| | - Satit Pandhumas
- Chulabhorn Wildlife Breeding Center, Department of National Parks, Wildlife and Plant Conservation, Sisaket 33140, Thailand
| | - Kiattisak Kaewsen
- Banglamung Wildlife Breeding Center, Department of National Parks, Wildlife and Plant Conservation, Chonburi 20150, Thailand
| | - Sekkarin Ploypetch
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Sujin Sirisawadi
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nanthida Kunnasut
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panat Anuracpreeda
- Parasitology Research Laboratory, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Amaya Watthanadirek-Wijidwong
- Parasitology Research Laboratory, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Gunnaporn Suriyaphol
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
6
|
Cruz KP, Petersen ALOA, Amorim MF, Pinho AGSF, Palma LC, Dantas DAS, Silveira MRG, Silva CSA, Cordeiro ALJ, Oliveira IG, Pita GB, Souza BCA, Bomfim GC, Brodskyn CI, Fraga DBM, Lima IS, de_Santana MBR, Teixeira HMP, de_Menezes JPB, Santos WLC, Veras PST. Intraperitoneal Administration of 17-DMAG as an Effective Treatment against Leishmania braziliensis Infection in BALB/c Mice: A Preclinical Study. Pathogens 2024; 13:630. [PMID: 39204231 PMCID: PMC11357173 DOI: 10.3390/pathogens13080630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Leishmaniasis is a significant global public health issue that is caused by parasites from Leishmania genus. With limited treatment options and rising drug resistance, there is a pressing need for new therapeutic approaches. Molecular chaperones, particularly Hsp90, play a crucial role in parasite biology and are emerging as promising targets for drug development. OBJECTIVE This study evaluates the efficacy of 17-DMAG in treating BALB/c mice from cutaneous leishmaniasis through in vitro and in vivo approaches. MATERIALS AND METHODS We assessed 17-DMAG's cytotoxic effect on bone marrow-derived macrophages (BMMΦ) and its effects against L. braziliensis promastigotes and intracellular amastigotes. Additionally, we tested the compound's efficacy in BALB/c mice infected with L. braziliensis via intraperitoneal administration to evaluate the reduction in lesion size and the decrease in parasite load in the ears and lymph nodes of infected animals. RESULTS 17-DMAG showed selective toxicity [selective index = 432) towards Leishmania amastigotes, causing minimal damage to host cells. The treatment significantly reduced lesion sizes in mice and resulted in parasite clearance from ears and lymph nodes. It also diminished inflammatory responses and reduced the release of pro-inflammatory cytokines (IL-6, IFN-γ, TNF) and the regulatory cytokine IL-10, underscoring its dual leishmanicidal and anti-inflammatory properties. CONCLUSIONS Our findings confirm the potential of 17-DMAG as a viable treatment for cutaneous leishmaniasis and support further research into its mechanisms and potential applications against other infectious diseases.
Collapse
Affiliation(s)
- Kercia P. Cruz
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Antonio L. O. A. Petersen
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
- Baiano Federal Institute of Education, Science and Technology—Santa Inês Campus, BR 420, Santa Inês Road, Rural Zone, Ubaíra 45320-000, Bahia, Brazil
| | - Marina F. Amorim
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Alan G. S. F. Pinho
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Luana C. Palma
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Diana A. S. Dantas
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Mariana R. G. Silveira
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Carine S. A. Silva
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Ana Luiza J. Cordeiro
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Izabella G. Oliveira
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Gabriella B. Pita
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Bianca C. A. Souza
- Laboratory of Structural and Molecular Pathology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (B.C.A.S.); (I.S.L.); (W.L.C.S.)
| | - Gilberto C. Bomfim
- Laboratory of Population Genetics and Molecular Evolution, Biology Institute, Federal University of Bahia, Salvador 40170-110, Bahia, Brazil;
| | - Cláudia I. Brodskyn
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Deborah B. M. Fraga
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
- Department of Preventive Veterinary Medicine and Animal Production, School of Veterinary Medicine and Animal Science, Federal University of Bahia, Salvador 40170-110, Bahia, Brazil
- National Institute of Science and Technology of Tropical Diseases (INCT-DT), National Council for Scientific Research and Development (CNPq)
| | - Isadora S. Lima
- Laboratory of Structural and Molecular Pathology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (B.C.A.S.); (I.S.L.); (W.L.C.S.)
| | - Maria B. R. de_Santana
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Helena M. P. Teixeira
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Juliana P. B. de_Menezes
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
| | - Washington L. C. Santos
- Laboratory of Structural and Molecular Pathology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (B.C.A.S.); (I.S.L.); (W.L.C.S.)
- Department of Pathology and Forensic Medicine, Bahia Medical School, Federal University of Bahia, Salvador 40110-906, Bahia, Brazil
| | - Patrícia S. T. Veras
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador 40296-710, Bahia, Brazil; (K.P.C.); (A.L.O.A.P.); (M.F.A.); (A.G.S.F.P.); (L.C.P.); (D.A.S.D.); (M.R.G.S.); (C.S.A.S.); (A.L.J.C.); (I.G.O.); (G.B.P.); (C.I.B.); (D.B.M.F.); (M.B.R.d.); (H.M.P.T.); (J.P.B.d.)
- National Institute of Science and Technology of Tropical Diseases (INCT-DT), National Council for Scientific Research and Development (CNPq)
| |
Collapse
|
7
|
Vashishtha S, Thakur S, Singh J, Adhana S, Kundu B. Evolutionarily conserved heat shock protein, HtpX, as an adjunct target against antibiotic-resistant Neisseria gonorrhoeae. J Cell Biochem 2023; 124:1516-1529. [PMID: 37566682 DOI: 10.1002/jcb.30461] [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: 02/26/2023] [Revised: 07/09/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
The emergence of multiple drug resistance and extreme drug resistance pathogens necessitates the continuous evaluation of the pathogenic genome to identify conserved molecular targets and their respective inhibitors. In this study, we mapped the global mutational landscape of Neisseria gonorrhoeae (an intracellular pathogen notoriously known to cause the sexually transmitted disease gonorrhoea). We identified highly variable amino acid positions in the antibiotic target genes like the penA, ponA, 23s rRNA, rpoB, gyrA, parC, mtrR and porB. Some variations are directly reported to confer resistance to the currently used front-line drugs like ceftriaxone, cefixime, azithromycin and ciprofloxacin. Further, by whole genome comparison and Shannon entropy analysis, we identified a completely conserved protein HtpX in the drug-resistant as well as susceptible isolates of N. gonorrhoeae (NgHtpX). Comparison with the only available information of Escherichia coli HtpX suggested it to be a transmembrane metalloprotease having a role in stress response. The critical zinc-binding residue of NgHtpX was mapped to E141. By applying composite high throughput screening followed by MD simulations, we identified pemirolast and thalidomide as high-energy binding ligands of NgHtpX. Following cloning and expression of the purified metal-binding domain of NgHtpX (NgHtpXd), its Zn2+ -binding (Kd = 0.4 µM) and drug-binding (pemirolast, Kd = 3.47 µM; and thalidomide, Kd = 1.04 µM) potentials were determined using in-vitro fluorescence quenching experiment. When tested on N. gonorrhoeae cultures, both the ligands imposed a dose-dependent reduction in viability. Overall, our results provide high entropy positions in the targets of presently used antibiotics, which can be further explored to understand the AMR mechanism. Additionally, HtpX and its specific inhibitors identified can be utilised effectively in managing gonococcal infections.
Collapse
Affiliation(s)
- Shubham Vashishtha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Sheetal Thakur
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Jasdeep Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Sujata Adhana
- Department of Biomedical Sciences, Bhaskaracharya College of Applied Sciences, University of Delhi, New Delhi, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
8
|
Bohmer MJ, Wang J, Istvan ES, Luth MR, Collins JE, Huttlin EL, Wang L, Mittal N, Hao M, Kwiatkowski NP, Gygi SP, Chakrabarti R, Deng X, Goldberg DE, Winzeler EA, Gray NS, Chakrabarti D. Human Polo-like Kinase Inhibitors as Antiplasmodials. ACS Infect Dis 2023; 9:1004-1021. [PMID: 36919909 PMCID: PMC10106425 DOI: 10.1021/acsinfecdis.3c00025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Protein kinases have proven to be a very productive class of therapeutic targets, and over 90 inhibitors are currently in clinical use primarily for the treatment of cancer. Repurposing these inhibitors as antimalarials could provide an accelerated path to drug development. In this study, we identified BI-2536, a known potent human polo-like kinase 1 inhibitor, with low nanomolar antiplasmodial activity. Screening of additional PLK1 inhibitors revealed further antiplasmodial candidates despite the lack of an obvious orthologue of PLKs in Plasmodium. A subset of these inhibitors was profiled for their in vitro killing profile, and commonalities between the killing rate and inhibition of nuclear replication were noted. A kinase panel screen identified PfNEK3 as a shared target of these PLK1 inhibitors; however, phosphoproteome analysis confirmed distinct signaling pathways were disrupted by two structurally distinct inhibitors, suggesting PfNEK3 may not be the sole target. Genomic analysis of BI-2536-resistant parasites revealed mutations in genes associated with the starvation-induced stress response, suggesting BI-2536 may also inhibit an aminoacyl-tRNA synthetase.
Collapse
Affiliation(s)
- Monica J Bohmer
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida 32826, United States
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biolo gy, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Eva S Istvan
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Madeline R Luth
- Department of Pediatrics, School of Medicine, University California, San Diego, La Jolla, California 92093, United States
| | - Jennifer E Collins
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida 32826, United States
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Lushun Wang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Nimisha Mittal
- Department of Pediatrics, School of Medicine, University California, San Diego, La Jolla, California 92093, United States
| | - Mingfeng Hao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biolo gy, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Nicholas P Kwiatkowski
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biolo gy, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ratna Chakrabarti
- Division of Cancer Research, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida 32826, United States
| | - Xianming Deng
- School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University California, San Diego, La Jolla, California 92093, United States
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Debopam Chakrabarti
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida 32826, United States
| |
Collapse
|
9
|
Tassone G, Mazzorana M, Pozzi C. Structural Basis of Parasitic HSP90 ATPase Inhibition by Small Molecules. Pharmaceuticals (Basel) 2022; 15:1341. [PMID: 36355513 PMCID: PMC9692773 DOI: 10.3390/ph15111341] [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: 09/23/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 09/10/2024] Open
Abstract
Protozoan parasites are responsible for several harmful and widespread human diseases that cause high morbidity and mortality. Currently available treatments have serious limitations due to poor efficiency, strong adverse effects, and high cost. Hence, the identification of new targets and the development of specific drug therapies against parasitic diseases are urgent needs. Heat shock protein 90 (HSP90) is an ATP-dependent molecular chaperone that plays a key role in parasite survival during the various differentiation stages, spread over the vector insect and the human host, which they undergo during their life cycle. The N-terminal domain (NTD) of HSP90, containing the main determinants for ATPase activity, represents the most druggable domain for inhibitor targeting. The molecules investigated on parasite HSP90 are mainly developed from known inhibitors of the human counterpart, and they have strong limitations due to selectivity issues, accounting for the high conservation of the ATP-binding site between the parasite and human proteins. The current review highlights the recent structural progress made to support the rational design of new molecules able to effectively block the chaperone activity of parasite HSP90.
Collapse
Affiliation(s)
- Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Marco Mazzorana
- Diamond Light Source Ltd., Diamond House, Harwell Science & Innovation Campus, Didcot OX11 0DE, UK
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
10
|
The Leishmania donovani Ortholog of the Glycosylphosphatidylinositol Anchor Biosynthesis Cofactor PBN1 Is Essential for Host Infection. mBio 2022; 13:e0043322. [PMID: 35420475 PMCID: PMC9239262 DOI: 10.1128/mbio.00433-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Visceral leishmaniasis is a deadly infectious disease caused by Leishmania donovani, a kinetoplastid parasite for which no licensed vaccine is available. To identify potential vaccine candidates, we systematically identified genes encoding putative cell surface and secreted proteins essential for parasite viability and host infection. We identified a protein encoded by LdBPK_061160 which, when ablated, resulted in a remarkable increase in parasite adhesion to tissue culture flasks. Here, we show that this phenotype is caused by the loss of glycosylphosphatidylinositol (GPI)-anchored surface molecules and that LdBPK_061160 encodes a noncatalytic component of the L. donovani GPI-mannosyltransferase I (GPI-MT I) complex. GPI-anchored surface molecules were rescued in the LdBPK_061160 mutant by the ectopic expression of both human genes PIG-X and PIG-M, but neither gene could complement the phenotype alone. From further sequence comparisons, we conclude that LdBPK_061160 is the functional orthologue of yeast PBN1 and mammalian PIG-X, which encode the noncatalytic subunits of their respective GPI-MT I complexes, and we assign LdBPK_061160 as LdPBN1. The LdPBN1 mutants could not establish a visceral infection in mice, a phenotype that was rescued by constitutive expression of LdPBN1. Although mice infected with the null mutant did not develop an infection, exposure to these parasites provided significant protection against subsequent infection with a virulent strain. In summary, we have identified the orthologue of the PBN1/PIG-X noncatalytic subunit of GPI-MT I in trypanosomatids, shown that it is essential for infection in a murine model of visceral leishmaniasis, and demonstrated that the LdPBN1 mutant shows promise for the development of an attenuated live vaccine.
Collapse
|
11
|
Stofberg ML, Caillet C, de Villiers M, Zininga T. Inhibitors of the Plasmodium falciparum Hsp90 towards Selective Antimalarial Drug Design: The Past, Present and Future. Cells 2021; 10:2849. [PMID: 34831072 PMCID: PMC8616389 DOI: 10.3390/cells10112849] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria is still one of the major killer parasitic diseases in tropical settings, posing a public health threat. The development of antimalarial drug resistance is reversing the gains made in attempts to control the disease. The parasite leads a complex life cycle that has adapted to outwit almost all known antimalarial drugs to date, including the first line of treatment, artesunate. There is a high unmet need to develop new strategies and identify novel therapeutics to reverse antimalarial drug resistance development. Among the strategies, here we focus and discuss the merits of the development of antimalarials targeting the Heat shock protein 90 (Hsp90) due to the central role it plays in protein quality control.
Collapse
Affiliation(s)
| | | | | | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa; (M.L.S.); (C.C.); (M.d.V.)
| |
Collapse
|
12
|
Tabassum W, Singh P, Suthram N, Bhattacharyya S, Bhattacharyya MK. Synergistic Action between PfHsp90 Inhibitor and PfRad51 Inhibitor Induces Elevated DNA Damage Sensitivity in the Malaria Parasite. Antimicrob Agents Chemother 2021; 65:e0045721. [PMID: 34097485 PMCID: PMC8370194 DOI: 10.1128/aac.00457-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/19/2021] [Indexed: 12/18/2022] Open
Abstract
The DNA recombinase Rad51 from the human malaria parasite Plasmodium falciparum has emerged as a potential drug target due to its central role in the homologous recombination (HR)-mediated double-strand break (DSB) repair pathway. Inhibition of the ATPase and strand exchange activity of P. falciparum Rad51 (PfRad51) by a small-molecule inhibitor, B02 [3-(phenylmethyl)-2-[(1E)-2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone], renders the parasite more sensitive to genotoxic agents. Here, we investigated whether the inhibition of the molecular chaperone PfHsp90 potentiates the antimalarial action of B02. We found that the PfHsp90 inhibitor 17-AAG [17-(allylamino)-17-demethoxygeldanamycin] exhibits strong synergism with B02 in both drug-sensitive (strain 3D7) and multidrug-resistant (strain Dd2) P. falciparum parasites. 17-AAG causes a greater than 200-fold decrease in the half-maximal inhibitory concentration (IC50) of B02 in 3D7 parasites. Our results provide mechanistic insights into such profound synergism between 17-AAG and B02. We report that PfHsp90 physically interacts with PfRad51 and promotes the UV irradiation-induced DNA repair activity of PfRad51 by controlling its stability. We find that 17-AAG reduces PfRad51 protein levels by accelerating proteasomal degradation. Consequently, PfHsp90 inhibition renders the parasites more susceptible to the potent DNA-damaging agent methyl methanesulfonate (MMS) in a dose-dependent manner. Thus, our study provides a rationale for targeting PfHsp90 along with the recombinase PfRad51 for controlling malaria propagation.
Collapse
Affiliation(s)
- Wahida Tabassum
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Priyanka Singh
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Niranjan Suthram
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sunanda Bhattacharyya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | |
Collapse
|
13
|
Martinez-Peinado N, Martori C, Cortes-Serra N, Sherman J, Rodriguez A, Gascon J, Alberola J, Pinazo MJ, Rodriguez-Cortes A, Alonso-Padilla J. Anti- Trypanosoma cruzi Activity of Metabolism Modifier Compounds. Int J Mol Sci 2021; 22:ijms22020688. [PMID: 33445756 PMCID: PMC7828178 DOI: 10.3390/ijms22020688] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/28/2020] [Accepted: 01/09/2021] [Indexed: 12/18/2022] Open
Abstract
Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and affects over 6 million people worldwide. Development of new drugs to treat this disease remains a priority since those currently available have variable efficacy and frequent adverse effects, especially during the long regimens required for treating the chronic stage of the disease. T. cruzi modulates the host cell-metabolism to accommodate the cell cytosol into a favorable growth environment and acquire nutrients for its multiplication. In this study we evaluated the specific anti-T. cruzi activity of nine bio-energetic modulator compounds. Notably, we identified that 17-DMAG, which targets the ATP-binding site of heat shock protein 90 (Hsp90), has a very high (sub-micromolar range) selective inhibition of the parasite growth. This inhibitory effect was also highly potent (IC50 = 0.27 μmol L-1) against the amastigote intracellular replicative stage of the parasite. Moreover, molecular docking results suggest that 17-DMAG may bind T. cruzi Hsp90 homologue Hsp83 with good affinity. Evaluation in a mouse model of chronic T. cruzi infection did not show parasite growth inhibition, highlighting the difficulties encountered when going from in vitro assays onto preclinical drug developmental stages.
Collapse
Affiliation(s)
- Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, 08036 Barcelona, Spain; (N.M.-P.); (N.C.-S.); (J.G.); (M.-J.P.)
| | - Clara Martori
- Department of Pharmacology, Toxicology, and Therapeutics, Veterinary Faculty, Autonomous University of Barcelona, 08193 Bellaterra, Spain; (C.M.); (J.A.)
| | - Nuria Cortes-Serra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, 08036 Barcelona, Spain; (N.M.-P.); (N.C.-S.); (J.G.); (M.-J.P.)
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA; (J.S.); (A.R.)
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA; (J.S.); (A.R.)
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, 08036 Barcelona, Spain; (N.M.-P.); (N.C.-S.); (J.G.); (M.-J.P.)
| | - Jordi Alberola
- Department of Pharmacology, Toxicology, and Therapeutics, Veterinary Faculty, Autonomous University of Barcelona, 08193 Bellaterra, Spain; (C.M.); (J.A.)
| | - Maria-Jesus Pinazo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, 08036 Barcelona, Spain; (N.M.-P.); (N.C.-S.); (J.G.); (M.-J.P.)
| | - Alheli Rodriguez-Cortes
- Department of Pharmacology, Toxicology, and Therapeutics, Veterinary Faculty, Autonomous University of Barcelona, 08193 Bellaterra, Spain; (C.M.); (J.A.)
- Correspondence: (A.R.-C.); (J.A.-P.); Tel.: +34-935812062 (A.R.-C.); +34-932275400 (J.A.-P.)
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, 08036 Barcelona, Spain; (N.M.-P.); (N.C.-S.); (J.G.); (M.-J.P.)
- Correspondence: (A.R.-C.); (J.A.-P.); Tel.: +34-935812062 (A.R.-C.); +34-932275400 (J.A.-P.)
| |
Collapse
|
14
|
Moon SJ, Jeong BC, Kim HJ, Lim JE, Kim HJ, Kwon GY, Jackman JA, Kim JH. Bruceantin targets HSP90 to overcome resistance to hormone therapy in castration-resistant prostate cancer. Am J Cancer Res 2021; 11:958-973. [PMID: 33391515 PMCID: PMC7738850 DOI: 10.7150/thno.51478] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/20/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: Aberrant androgen receptor (AR) signaling via full-length AR (AR-FL) and constitutively active AR variant 7 (AR-V7) plays a key role in the development of castration-resistant prostate cancer (CRPC) and resistance to hormone therapies. Simultaneous targeting of AR-FL and AR-V7 may be a promising strategy to overcome resistance to hormone therapy. This study aimed to identify novel drug candidates co-targeting AR-FL and AR-V7 activities and elucidate their molecular mechanism of anti-CRPC activities. Methods: Using a CRPC cell-based reporter assay system, we screened a small library of antimalarial agents to explore the possibility of repositioning them for CRPC treatment and identified bruceantin (BCT) as a potent anti-CRPC drug candidate. A series of cell-based, molecular, biochemical, and in vivo approaches were performed to evaluate the therapeutic potential and molecular mechanism of BCT in CRPC. These approaches include reporter gene assays, cell proliferation, RNA-seq, qRT-PCR, mouse xenografts, co-immunoprecipitation, GST pull-down, immobilized BCT pull-down, molecular modeling, and bioinformatic analyses. Results: We identified BCT as a highly potent inhibitor co-targeting AR-FL and AR-V7 activity. BCT inhibits the transcriptional activity of AR-FL/AR-V7 and downregulates their target genes in CRPC cells. In addition, BCT efficiently suppresses tumor growth and metastasis of CRPC cells. Mechanistically, BCT disrupts the interaction of HSP90 with AR-FL/AR-V7 by directly binding to HSP90 and inhibits HSP90 chaperone function, leading to degradation of AR-FL/AR-V7 through the ubiquitin-proteasome system. Clinically, HSP90 expression is upregulated and correlated with AR/AR-V7 levels in CRPC. Conclusion: Our findings suggest that BCT could serve as a promising therapeutic candidate against CRPC and highlight the potential benefit of targeting AR-FL/AR-V7-HSP90 axis to overcome resistance caused by aberrant AR-FL/AR-V7 signaling.
Collapse
|
15
|
Elmehy DA, Ismail HI, Soliman NA, Amer BS, Elkaliny HH, El-Ebiary AA, Gamea GA. Oxidative stress mediated apoptotic potential of mefloquine on experimental trichinellosis. Acta Trop 2021; 213:105760. [PMID: 33221280 DOI: 10.1016/j.actatropica.2020.105760] [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] [Received: 02/10/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 10/23/2022]
Abstract
Conventional anthelmintics such as albendazole could not achieve complete cure of trichinellosis till now. The antimalarial mefloquine mediates oxidative stress and disrupts lysosomal functions leading to cell death. Therefore, the aim of this work was to investigate the effect of mefloquine on experimental acute and chronic trichinellosis and to clarify the possible mechanisms of such effects. Mice were divided into four groups; Group I: Uninfected untreated control (20 mice); Group II: Infected untreated control (40 mice); Group III: infected and treated with albendazole (400 mg/kg) (40 mice); Group IV: infected and treated with mefloquine (300 mg/kg) (40 mice). All infected treated groups were equally subdivided into 2 subgroups; (a) treated on the 2nd day post infection (dpi) for 3 days, (b) treated on the 35th dpi for 5 days. Parasitological adults and larvae counting besides immunohistopathological examination of intestines and muscles were done. Biochemical assay of oxidant/antioxidant status, apoptotic, cytoprotective and inflammatory biomarkers in intestinal and muscle homogenates were achieved. Results showed that both albendazole and mefloquine significantly reduced adults and larvae counts with higher efficacy of albendazole in the intestinal phase and superiority of mefloquine in the muscle phase. The superiority of mefloquine was indicated by increased inflammatory immune infiltration and decreased anti-apoptotic immunohistochemical markers expression in both jejunal and muscle tissues. Biochemically, mefloquine treatment showed highly significant oxidative, apoptotic and inflammatory effects. So, our results suggest that mefloquine might be a superior treatment for chronic trichinellosis.
Collapse
|
16
|
Zininga T, Shonhai A. Small Molecule Inhibitors Targeting the Heat Shock Protein System of Human Obligate Protozoan Parasites. Int J Mol Sci 2019; 20:E5930. [PMID: 31775392 PMCID: PMC6929125 DOI: 10.3390/ijms20235930] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Obligate protozoan parasites of the kinetoplastids and apicomplexa infect human cells to complete their life cycles. Some of the members of these groups of parasites develop in at least two systems, the human host and the insect vector. Survival under the varied physiological conditions associated with the human host and in the arthropod vectors requires the parasites to modulate their metabolic complement in order to meet the prevailing conditions. One of the key features of these parasites essential for their survival and host infectivity is timely expression of various proteins. Even more importantly is the need to keep their proteome functional by maintaining its functional capabilities in the wake of physiological changes and host immune responses. For this reason, molecular chaperones (also called heat shock proteins)-whose role is to facilitate proteostasis-play an important role in the survival of these parasites. Heat shock protein 90 (Hsp90) and Hsp70 are prominent molecular chaperones that are generally induced in response to physiological stress. Both Hsp90 and Hsp70 members are functionally regulated by nucleotides. In addition, Hsp70 and Hsp90 cooperate to facilitate folding of some key proteins implicated in cellular development. In addition, Hsp90 and Hsp70 individually interact with other accessory proteins (co-chaperones) that regulate their functions. The dependency of these proteins on nucleotide for their chaperone function presents an Achille's heel, as inhibitors that mimic ATP are amongst potential therapeutic agents targeting their function in obligate intracellular human parasites. Most of the promising small molecule inhibitors of parasitic heat shock proteins are either antibiotics or anticancer agents, whose repurposing against parasitic infections holds prospects. Both cancer cells and obligate human parasites depend upon a robust protein quality control system to ensure their survival, and hence, both employ a competent heat shock machinery to this end. Furthermore, some inhibitors that target chaperone and co-chaperone networks also offer promising prospects as antiparasitic agents. The current review highlights the progress made so far in design and application of small molecule inhibitors against obligate intracellular human parasites of the kinetoplastida and apicomplexan kingdoms.
Collapse
Affiliation(s)
| | - Addmore Shonhai
- Department of Biochemistry, School of Mathematical and Natural Sciences, University of Venda, Thohoyandou 0950, South Africa;
| |
Collapse
|
17
|
A docking-based structural analysis of geldanamycin-derived inhibitor binding to human or Leishmania Hsp90. Sci Rep 2019; 9:14756. [PMID: 31611575 PMCID: PMC6791876 DOI: 10.1038/s41598-019-51239-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Leishmaniasis is a neglected disease that affects millions of individuals around the world. Regardless of clinical form, treatment is based primarily on the use of pentavalent antimonials. However, such treatments are prolonged and present intense side effects, which lead to patient abandonment in many cases. The search for chemotherapeutic alternatives has become a priority. Heat Shock Protein 90 (Hsp90) inhibitors have recently come under investigation due to antiparasitic activity in Plasmodium sp., Trypanosoma sp. and Leishmania sp. Some of these inhibitors, such as geldanamycin and its analogs, 17-AAG and 17-DMAG, bind directly to Hsp90, thereby inhibiting its activity. Previous studies have demonstrated that different parasite species are more susceptible to some of these inhibitors than host cells. We hypothesized that this increased susceptibility may be due to differences in binding of Hsp90 inhibitors to Leishmania protein compared to host protein. Based on the results of the in silico approach used in the present study, we propose that geldanamycin, 17-AAG and 17-DMAG present an increased tendency to bind to the N-terminal domain of Leishmania amazonensis Hsp83 in comparison to human Hsp90. This could be partially explained by differences in intermolecular interactions between each of these inhibitors and Hsp83 or Hsp90. The present findings demonstrate potential for the use of these inhibitors in the context of anti-Leishmania therapy.
Collapse
|
18
|
Engel JA, Norris EL, Gilson P, Przyborski J, Shonhai A, Blatch GL, Skinner-Adams TS, Gorman J, Headlam M, Andrews KT. Proteomic analysis of Plasmodium falciparum histone deacetylase 1 complex proteins. Exp Parasitol 2019; 198:7-16. [PMID: 30682336 DOI: 10.1016/j.exppara.2019.01.008] [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] [Received: 08/21/2018] [Revised: 12/01/2018] [Accepted: 01/20/2019] [Indexed: 01/12/2023]
Abstract
Plasmodium falciparum histone deacetylases (PfHDACs) are an important class of epigenetic regulators that alter protein lysine acetylation, contributing to regulation of gene expression and normal parasite growth and development. PfHDACs are therefore under investigation as drug targets for malaria. Despite this, our understanding of the biological roles of these enzymes is only just beginning to emerge. In higher eukaryotes, HDACs function as part of multi-protein complexes and act on both histone and non-histone substrates. Here, we present a proteomics analysis of PfHDAC1 immunoprecipitates, identifying 26 putative P. falciparum complex proteins in trophozoite-stage asexual intraerythrocytic parasites. The co-migration of two of these (P. falciparum heat shock proteins 70-1 and 90) with PfHDAC1 was validated using Blue Native PAGE combined with Western blot. These data provide a snapshot of possible PfHDAC1 interactions and a starting point for future studies focused on elucidating the broader function of PfHDACs in Plasmodium parasites.
Collapse
Affiliation(s)
- Jessica A Engel
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Emma L Norris
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Paul Gilson
- Burnet Institute, Monash University, Victoria, Australia
| | - Jude Przyborski
- Centre of Infectious Diseases, Parasitology, University Hospital Heidelberg, Germany
| | - Addmore Shonhai
- Biochemistry Department, University of Venda, Thohoyandou, South Africa
| | - Gregory L Blatch
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Tina S Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Jeffrey Gorman
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | | | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia.
| |
Collapse
|
19
|
Quiliano M, Pabón A, Moles E, Bonilla-Ramirez L, Fabing I, Fong KY, Nieto-Aco DA, Wright DW, Pizarro JC, Vettorazzi A, López de Cerain A, Deharo E, Fernández-Busquets X, Garavito G, Aldana I, Galiano S. Structure-activity relationship of new antimalarial 1-aryl-3-susbtituted propanol derivatives: Synthesis, preliminary toxicity profiling, parasite life cycle stage studies, target exploration, and targeted delivery. Eur J Med Chem 2018; 152:489-514. [PMID: 29754074 DOI: 10.1016/j.ejmech.2018.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 01/09/2023]
Abstract
Design, synthesis, structure-activity relationship, cytotoxicity studies, in silico drug-likeness, genotoxicity screening, and in vivo studies of new 1-aryl-3-substituted propanol derivatives led to the identification of nine compounds with promising in vitro (55, 56, 61, 64, 66, and 70-73) and in vivo (66 and 72) antimalarial profiles against Plasmodium falciparum and Plasmodium berghei. Compounds 55, 56, 61, 64, 66 and 70-73 exhibited potent antiplasmodial activity against chloroquine-resistant strain FCR-3 (IC50s < 0.28 μM), and compounds 55, 56, 64, 70, 71, and 72 showed potent biological activity in chloroquine-sensitive and multidrug-resistant strains (IC50s < 0.7 μM for 3D7, D6, FCR-3 and C235). All of these compounds share appropriate drug-likeness profiles and adequate selectivity indexes (77 < SI < 184) as well as lack genotoxicity. In vivo efficacy tests in a mouse model showed compounds 66 and 72 to be promising candidates as they exhibited significant parasitemia reductions of 96.4% and 80.4%, respectively. Additional studies such as liver stage and sporogony inhibition, target exploration of heat shock protein 90 of P. falciparum, targeted delivery by immunoliposomes, and enantiomer characterization were performed and strongly reinforce the hypothesis of 1-aryl-3-substituted propanol derivatives as promising antimalarial compounds.
Collapse
Affiliation(s)
- Miguel Quiliano
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - Adriana Pabón
- Grupo Malaria, Universidad de Antioquía, Medellín, Colombia
| | - Ernest Moles
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, 08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | | | - Isabelle Fabing
- Laboratoire de Synthese et Physicochimie de Molécules d'Intéret Biologique SPCMIB-UMR5068, CNRS - Université Paul Sabatier, 118, route de Narbonne, 31062, Toulouse Cedex 09, France
| | - Kim Y Fong
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - Diego A Nieto-Aco
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - David W Wright
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University USA; Vector-Borne Infectious Diseases Research Center, Tulane University USA
| | - Ariane Vettorazzi
- Universidad de Navarra, Facultad de Farmacia y Nutrición, Department of Pharmacology and Toxicology, Campus Universitario, 31008 Pamplona, Spain
| | - Adela López de Cerain
- Universidad de Navarra, Facultad de Farmacia y Nutrición, Department of Pharmacology and Toxicology, Campus Universitario, 31008 Pamplona, Spain
| | - Eric Deharo
- UMR 152 PHARMA-DEV, Université Toulouse, IRD, UPS, 31062, Toulouse, France
| | - Xavier Fernández-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, 08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Giovanny Garavito
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Departamento de Farmacia (DFUNC), Grupo de investigación FaMeTra (Farmacología de la Medicina tradicional y popular), Carrera 30 45-03, Bogotá D.C., Colombia
| | - Ignacio Aldana
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - Silvia Galiano
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain.
| |
Collapse
|