1
|
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] [MESH Headings] [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
|
2
|
Wickramaratne AC, Wickner S, Kravats AN. Hsp90, a team player in protein quality control and the stress response in bacteria. Microbiol Mol Biol Rev 2024; 88:e0017622. [PMID: 38534118 PMCID: PMC11332350 DOI: 10.1128/mmbr.00176-22] [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: 03/28/2024] Open
Abstract
SUMMARYHeat shock protein 90 (Hsp90) participates in proteostasis by facilitating protein folding, activation, disaggregation, prevention of aggregation, degradation, and protection against degradation of various cellular proteins. It is highly conserved from bacteria to humans. In bacteria, protein remodeling by Hsp90 involves collaboration with the Hsp70 molecular chaperone and Hsp70 cochaperones. In eukaryotes, protein folding by Hsp90 is more complex and involves collaboration with many Hsp90 cochaperones as well as Hsp70 and Hsp70 cochaperones. This review focuses primarily on bacterial Hsp90 and highlights similarities and differences between bacterial and eukaryotic Hsp90. Seminal research findings that elucidate the structure and the mechanisms of protein folding, disaggregation, and reactivation promoted by Hsp90 are discussed. Understanding the mechanisms of bacterial Hsp90 will provide fundamental insight into the more complex eukaryotic chaperone systems.
Collapse
Affiliation(s)
- Anushka C. Wickramaratne
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sue Wickner
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrea N. Kravats
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio, USA
| |
Collapse
|
3
|
Marín M, López M, Gallego-Yerga L, Álvarez R, Peláez R. Experimental structure based drug design (SBDD) applications for anti-leishmanial drugs: A paradigm shift? Med Res Rev 2024; 44:1055-1120. [PMID: 38142308 DOI: 10.1002/med.22005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.
Collapse
Affiliation(s)
- Miguel Marín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Marta López
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
4
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [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] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
5
|
Tsutsumi H, Abe M, Uchida N, Takiguchi M, Yamasaki M. The role of heat shock protein 90 in the proliferation of Babesia gibsoni in vitro. Exp Parasitol 2023:108567. [PMID: 37308002 DOI: 10.1016/j.exppara.2023.108567] [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: 06/02/2022] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/14/2023]
Abstract
The present study investigated the role of heat shock protein 90 (HSP90) in the proliferation and survival of Babesia gibsoni in vitro. To detect the effect on the entry of B. gibsoni into host erythrocytes, the parasite was incubated with an antibody against B. gibsoni HSP90 (BgHSP90) for 24 hr. The results of this experiment demonstrated that both the incorporation of [3H]hypoxanthine into the nucleic acids of B. gibsoni and the number of parasites were not altered, indicating that an anti-BgHSP90 antibody did not directly inhibit the entry of the parasite into erythrocytes. Moreover, two HSP90 inhibitors, geldanamycin (GA) and tanespimycin (17-AAG), were used to evaluate the function of BgHSP90. GA and 17-AAG decreased both the incorporation of [3H]hypoxanthine and the number of infected erythrocytes, suggesting that BgHSP90 plays important roles in DNA synthesis and the proliferation of B. gibsoni. The effect of 17-AAG on the parasites was weaker than that of GA. Additionally, the effect of GA on the survival and superoxide generation of canine neutrophils was assessed. The survival of canine neutrophils was not affected. The superoxide generation was strongly suppressed by GA. This result indicated that GA inhibited the function of canine neutrophils. Additional studies are necessary to elucidate the role of BgHSP90 in the proliferation of the parasite.
Collapse
Affiliation(s)
- Hiroka Tsutsumi
- Laboratory of Veterinary Small Animal Internal Medicine, Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 202-8550, Japan
| | - Moeko Abe
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Naohiro Uchida
- Laboratory of Veterinary Small Animal Internal Medicine, Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 202-8550, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Masahiro Yamasaki
- Laboratory of Veterinary Small Animal Internal Medicine, Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 202-8550, Japan.
| |
Collapse
|
6
|
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
|
7
|
Jamabo M, Bentley SJ, Macucule-Tinga P, Tembo P, Edkins AL, Boshoff A. In silico analysis of the HSP90 chaperone system from the African trypanosome, Trypanosoma brucei. Front Mol Biosci 2022; 9:947078. [PMID: 36213128 PMCID: PMC9538636 DOI: 10.3389/fmolb.2022.947078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
African trypanosomiasis is a neglected tropical disease caused by Trypanosoma brucei (T. brucei) and spread by the tsetse fly in sub-Saharan Africa. The trypanosome relies on heat shock proteins for survival in the insect vector and mammalian host. Heat shock protein 90 (HSP90) plays a crucial role in the stress response at the cellular level. Inhibition of its interactions with chaperones and co-chaperones is being explored as a potential therapeutic target for numerous diseases. This study provides an in silico overview of HSP90 and its co-chaperones in both T. brucei brucei and T. brucei gambiense in relation to human and other trypanosomal species, including non-parasitic Bodo saltans and the insect infecting Crithidia fasciculata. A structural analysis of T. brucei HSP90 revealed differences in the orientation of the linker and C-terminal domain in comparison to human HSP90. Phylogenetic analysis displayed the T. brucei HSP90 proteins clustering into three distinct groups based on subcellular localizations, namely, cytosol, mitochondria, and endoplasmic reticulum. Syntenic analysis of cytosolic HSP90 genes revealed that T. b. brucei encoded for 10 tandem copies, while T. b. gambiense encoded for three tandem copies; Leishmania major (L. major) had the highest gene copy number with 17 tandem copies. The updated information on HSP90 from recently published proteomics on T. brucei was examined for different life cycle stages and subcellular localizations. The results show a difference between T. b. brucei and T. b. gambiense with T. b. brucei encoding a total of twelve putative HSP90 genes, while T. b. gambiense encodes five HSP90 genes. Eighteen putative co-chaperones were identified with one notable absence being cell division cycle 37 (Cdc37). These results provide an updated framework on approaching HSP90 and its interactions as drug targets in the African trypanosome.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
| | | | | | - Praise Tembo
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
| | - Adrienne Lesley Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, South Africa
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
- *Correspondence: Aileen Boshoff,
| |
Collapse
|
8
|
Kumari D, Mahajan S, Kour P, Singh K. Virulence factors of Leishmania parasite: Their paramount importance in unraveling novel vaccine candidates and therapeutic targets. Life Sci 2022; 306:120829. [PMID: 35872004 DOI: 10.1016/j.lfs.2022.120829] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a neglected tropical disease and remains a global concern for healthcare. It is caused by an opportunistic protozoan parasite belonging to the genus Leishmania and affects millions worldwide. This disease is mainly prevalent in tropical and subtropical regions and is associated with a high risk of public morbidity and mortality if left untreated. Transmission of this deadly disease is aggravated by the bite of female sand-fly vectors (Phlebotomus and Lutzomyia). With time, significant advancement in leishmaniasis-related research has been carried out to cope with the disease burden. Still, the Leishmania parasite has also co-evolved with its host and adapted successfully within the host's lethal milieu/environment. Thus, understanding and knowledge of various leishmanial virulence factors responsible for the parasitic infection are essential for exploring drug targets and vaccine candidates. The present review elucidates the importance of virulence factors in pathogenesis and summarizes the major leishmanial virulence molecules contributing to the parasitic infection during host-pathogen interaction. Furthermore, we have also elaborated on the potential contribution of leishmanial virulence proteins in developing vaccine candidates and exploring novel therapeutics against this parasitic disease. We aim to represent a clearer picture of parasite pathogenesis within the human host that can further aid in unraveling new strategies to fight against the deadly infection of leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shavi Mahajan
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Parampreet Kour
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
9
|
Cytosolic and Mitochondrial Hsp90 in Cytokinesis, Mitochondrial DNA Replication, and Drug Action in Trypanosoma brucei. Antimicrob Agents Chemother 2021; 65:e0063221. [PMID: 34424040 DOI: 10.1128/aac.00632-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Trypanosoma brucei subspecies cause African sleeping sickness in humans, an infection that is commonly fatal if not treated, and available therapies are limited. Previous studies have shown that heat shock protein 90 (Hsp90) inhibitors have potent and vivid activity against bloodstream-form trypanosomes. Hsp90s are phylogenetically conserved and essential catalysts that function at the crux of cell biology, where they ensure the proper folding of proteins and their assembly into multicomponent complexes. To assess the specificity of Hsp90 inhibitors and further define the role of Hsp90s in African trypanosomes, we used RNA interference (RNAi) to knock down cytosolic and mitochondrial Hsp90s (HSP83 and HSP84, respectively). Loss of either protein led to cell death, but the phenotypes were distinctly different. Depletion of cytosolic HSP83 closely mimicked the consequences of chemically depleting Hsp90 activity with inhibitor 17-AAG. In these cells, cytokinesis was severely disrupted, and segregation of the kinetoplast (the massive mitochondrial DNA structure unique to this family of eukaryotic pathogens) was impaired, leading to cells with abnormal kinetoplast DNA (kDNA) structures. Quite differently, knockdown of mitochondrial HSP84 did not impair cytokinesis but halted the initiation of new kDNA synthesis, generating cells without kDNA. These findings highlight the central role of Hsp90s in chaperoning cell cycle regulators in trypanosomes, reveal their unique function in kinetoplast replication, and reinforce their specificity and value as drug targets.
Collapse
|
10
|
17-AAG-Induced Activation of the Autophagic Pathway in Leishmania Is Associated with Parasite Death. Microorganisms 2021; 9:microorganisms9051089. [PMID: 34069389 PMCID: PMC8158731 DOI: 10.3390/microorganisms9051089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
The heat shock protein 90 (Hsp90) is thought to be an excellent drug target against parasitic diseases. The leishmanicidal effect of an Hsp90 inhibitor, 17-N-allylamino-17-demethoxygeldanamycin (17-AAG), was previously demonstrated in both in vitro and in vivo models of cutaneous leishmaniasis. Parasite death was shown to occur in association with severe ultrastructural alterations in Leishmania, suggestive of autophagic activation. We hypothesized that 17-AAG treatment results in the abnormal activation of the autophagic pathway, leading to parasite death. To elucidate this process, experiments were performed using transgenic parasites with GFP-ATG8-labelled autophagosomes. Mutant parasites treated with 17-AAG exhibited autophagosomes that did not entrap cargo, such as glycosomes, or fuse with lysosomes. ATG5-knockout (Δatg5) parasites, which are incapable of forming autophagosomes, demonstrated lower sensitivity to 17-AAG-induced cell death when compared to wild-type (WT) Leishmania, further supporting the role of autophagy in 17-AAG-induced cell death. In addition, Hsp90 inhibition resulted in greater accumulation of ubiquitylated proteins in both WT- and Δatg5-treated parasites compared to controls, in the absence of proteasome overload. In conjunction with previously described ultrastructural alterations, herein we present evidence that treatment with 17-AAG causes abnormal activation of the autophagic pathway, resulting in the formation of immature autophagosomes and, consequently, incidental parasite death.
Collapse
|
11
|
Heat Shock Proteins as the Druggable Targets in Leishmaniasis: Promises and Perils. Infect Immun 2021; 89:IAI.00559-20. [PMID: 33139381 DOI: 10.1128/iai.00559-20] [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: 12/24/2022] Open
Abstract
Leishmania, the causative agent of leishmaniasis, is an intracellular pathogen that thrives in the insect gut and mammalian macrophages to complete its life cycle. Apart from temperature difference (26 to 37°C), it encounters several harsh conditions, including oxidative stress, inflammatory reactions, and low pH. Heat shock proteins (HSPs) play essential roles in cell survival by strategically reprogramming cellular processes and signaling pathways. HSPs assist cells in multiple functions, including differentiation, adaptation, virulence, and persistence in the host cell. Due to cyclical epidemiological patterns, limited chemotherapeutic options, drug resistance, and the absence of a vaccine, control of leishmaniasis remains a far-fetched dream. The essential roles of HSPs in parasitic differentiation and virulence and increased expression in drug-resistant strains highlight their importance in combating the disease. In this review, we highlighted the diverse physiological importance of HSPs present in Leishmania, emphasizing their significance in disease pathogenesis. Subsequently, we assessed the potential of HSPs as a chemotherapeutic target and underlined the challenges associated with it. Furthermore, we have summarized a few ongoing drug discovery initiatives that need to be explored further to develop clinically successful chemotherapeutic agents in the future.
Collapse
|
12
|
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
|
13
|
Yamasaki M, Idaka N, Abe M, Takiguchi M. Reduced expression levels of heat shock protein 90 in a diminazene aceturate-resistant Babesia gibsoni isolate. Exp Parasitol 2020; 221:108050. [PMID: 33307095 DOI: 10.1016/j.exppara.2020.108050] [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: 03/24/2020] [Revised: 11/24/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Heat shock protein 90 (HSP90) is a molecular chaperon and an essential component for stage differentiation and intracellular growth inside the host cells of many protozoans. HSP90 of Babesia gibsoni (BgHSP90) was suggested to function in the development of diminazene aceturate (DA)-resistance. Therefore, we examined the expression level of BgHSP90 in a DA-resistant B. gibsoni isolate. Transcription of the BgHSP90 gene in the DA-resistant isolate and wild-type B. gibsoni was assessed by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). As a result, the copy number and relative amount of BgHSP90 transcripts in the DA-resistant isolate were significantly lower than those in the wild-type. Moreover, a rabbit anti-recombinant BgHSP90 antibody was developed, and the protein synthesis of BgHSP90 in the DA-resistant isolate was compared with that in the wild-type by Western blot analysis and indirect fluorescence assay. There was significantly less BgHSP90 protein than in the wild-type. Additionally, the relative intensity of BgHSP70 in DA-resistant isolate was also lower than that in the wild-type. This suggested that the expression of BgHSP90 and BgHSP70 in the DA-resistant B. gibsoni isolate was suppressed and that the reduced amount of BgHSP90 and BgHSP70 might cause the weak proliferation of the DA-resistant isolate. Further studies are necessary to elucidate the function of BgHSP90.
Collapse
Affiliation(s)
- Masahiro Yamasaki
- Laboratory of Veterinary Small Animal Internal Medicine, Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 202-8550, Japan.
| | - Natsuki Idaka
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Moeko Abe
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
14
|
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
|
15
|
Meyer KJ, Caton E, Shapiro TA. Model System Identifies Kinetic Driver of Hsp90 Inhibitor Activity against African Trypanosomes and Plasmodium falciparum. Antimicrob Agents Chemother 2018; 62:e00056-18. [PMID: 29866861 PMCID: PMC6105818 DOI: 10.1128/aac.00056-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/26/2018] [Indexed: 12/21/2022] Open
Abstract
Hsp90 inhibitors, well studied in the laboratory and clinic for antitumor indications, have promising activity against protozoan pathogens, including Trypanosoma brucei which causes African sleeping sickness, and the malaria parasite, Plasmodium falciparum To progress these experimental drugs toward clinical use, we adapted an in vitro dynamic hollow-fiber system and deployed artificial pharmacokinetics to discover the driver of their activity: either concentration or time. The activities of compounds from three major classes of Hsp90 inhibitors in development were evaluated against trypanosomes. In all circumstances, the activities of the tested Hsp90 inhibitors were concentration driven. By optimally deploying the drug to match its kinetic driver, the efficacy of a given dose was improved up to 5-fold, and maximal efficacy was achieved with a significantly lower drug exposure. The superiority of concentration-driven regimens was evident in vitro over several logs of drug exposure and was predictive of efficacy in a mouse model of African trypanosomiasis. In studies with P. falciparum, antimalarial activity was similarly concentration driven. This experimental strategy offers an expedient and versatile translational tool to assess the impact of pharmacokinetics on antiprotozoal activity. Knowing kinetic governance early in drug development provides an additional metric for judging lead compounds and allows the incisive design of animal efficacy studies.
Collapse
Affiliation(s)
- Kirsten J Meyer
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emily Caton
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Theresa A Shapiro
- Division of Clinical Pharmacology, Departments of Medicine and of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Guswanto A, Nugraha AB, Tuvshintulga B, Tayebwa DS, Rizk MA, Batiha GES, Gantuya S, Sivakumar T, Yokoyama N, Igarashi I. 17-DMAG inhibits the multiplication of several Babesia species and Theileria equi on in vitro cultures, and Babesia microti in mice. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2018; 8:104-111. [PMID: 29499568 PMCID: PMC6114103 DOI: 10.1016/j.ijpddr.2018.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 01/20/2023]
Abstract
Heat shock protein 90 (Hsp90) is a chaperone protein that stabilizes cells during stress or non-stress responses. Previous reports have shown that Hsp90 is a potential drug target to suppress the multiplication of several protozoan parasites. In this study, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), an Hsp90 inhibitor, was evaluated for its inhibitory effect on five in vitro cultures of Babesia and Theileria species, including B. bovis, B. bigemina, B. divergens, B. caballi, and T. equi, and on the multiplication of a B. microti–infected mouse model. 17-DMAG showed the inhibitory effect in all of the species tested. The half maximum inhibition concentration (IC50) of 17-DMAG on B. bovis, B. bigemina, B. divergens, B. caballi, and T. equi was 77.6 ± 2.9, 62.4 ± 1.9, 183.8 ± 3.2, 88.5 ± 9.6, and 307.7 ± 7.2 nM, respectively. The toxicity assay on MDBK and NIH/3T3 cell lines showed that 17-DMAG affected the viability of cells with an IC50 of 15.5 ± 4 and 8.8 ± 2 μM, respectively. Since the IC50s were much lower on the parasites than on the host cell lines, the selectivity index were high for all tested species. Furthermore, the two-drug combination of 17-DMAG with diminazene aceturate (DA) and atovaquone (AV) showed synergism or addition on in vitro cultures of Babesia and Theileria parasites. In the mouse model, 17-DMAG at a concentration of 30 mg/kg BW effectively inhibited the multiplication of B. microti. Moreover, if combined with DA or AV, 17-DMAG showed a comparable inhibition at the half dose. Taken together, these results indicate that 17-DMAG is a potent drug for treating piroplamosis. The data warrant further evaluation of 17-DMAG as an antibabesial drug and as an option in combination with atovaquone for the treatment of human babesiosis. 17-DMAG inhibits the in vitro multiplication of Babesia and Theileria parasites. Combination of 17-DMAG with diminazene aceturate or atovaquone were also effective. 17-DMAG also inhibits the multiplication of B. microti in mice. 17-DMAG is a new treatment option for babesiosis in animal and human.
Collapse
Affiliation(s)
- Azirwan Guswanto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan; Balai Veteriner Subang (DIC Subang), Jl. Terusan Garuda 33/11 Blok Werasari Dangdeur, Subang, Jawa Barat 41212, Indonesia.
| | - Arifin Budiman Nugraha
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan; Department of Animal Infectious Diseases and Veterinary Public Health, Faculty of Veterinary Medicine, Bogor Agricultural University, Jl. Agatis, Kampus IPB Dramaga, Bogor, Indonesia.
| | - Bumduuren Tuvshintulga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| | - Dickson Stuart Tayebwa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| | - Mohamed Abdo Rizk
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan; Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Gaber El-Saber Batiha
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan; Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Al-Beheira, 22511, Egypt.
| | - Sambuu Gantuya
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| | - Thillaiampalam Sivakumar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-13 Inada-cho, Obihiro 080-8555, Japan.
| |
Collapse
|
17
|
Berninger M, Schmidt I, Ponte-Sucre A, Holzgrabe U. Novel lead compounds in pre-clinical development against African sleeping sickness. MEDCHEMCOMM 2017; 8:1872-1890. [PMID: 30108710 PMCID: PMC6072528 DOI: 10.1039/c7md00280g] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/29/2017] [Indexed: 01/21/2023]
Abstract
Human African trypanosomiasis (HAT), also known as African sleeping sickness, is caused by parasitic protozoa of the genus Trypanosoma. As the disease progresses, the parasites cross the blood brain barrier and are lethal for the patients if the disease is left untreated. Current therapies suffer from several drawbacks due to e.g. toxicity of the respective compounds or resistance to approved antitrypanosomal drugs. In this review, the different strategies of drug development against HAT are considered, namely the target-based approach, the phenotypic high throughput screening and the drug repurposing strategy. The most promising compounds emerging from these approaches entering an in vivo evaluation are mentioned herein. Of note, it may turn out to be difficult to confirm in vitro activity in an animal model of infection; however, possible reasons for the missing efficacy in unsuccessful in vivo studies are discussed.
Collapse
Affiliation(s)
- Michael Berninger
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| | - Ines Schmidt
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology , Institute of Experimental Medicine , Luis Razetti School of Medicine , Faculty of Medicine , Universidad Central de Venezuela Caracas , Venezuela . Tel: +0931 31 85461
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| |
Collapse
|
18
|
Kanwar A, Eduful BJ, Barbeto L, Carletti Bonomo P, Lemus A, Vesely BA, Mutka TS, Azhari A, Kyle DE, Leahy JW. Synthesis and Activity of a New Series of Antileishmanial Agents. ACS Med Chem Lett 2017; 8:797-801. [PMID: 28835791 DOI: 10.1021/acsmedchemlett.7b00039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/31/2017] [Indexed: 11/30/2022] Open
Abstract
We have determined that tetrahydroindazoles such as 1 show potent activity against Leishmania donovani, the causative agent of leishmaniasis. While the Hsp90 activity and anticancer properties of 1 have previously been explored, we present here our efforts to optimize their activity against L. donovani via the synthesis of novel analogues designed to probe the hydrophobic pocket of the protozoan Hsp90 orthologue, specifically through the auspices of functionalization of an amine embedded into the scaffold.
Collapse
Affiliation(s)
- Ankush Kanwar
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Benjamin J. Eduful
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Linda Barbeto
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Piero Carletti Bonomo
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Andrea Lemus
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Brian A. Vesely
- Department
of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, Florida 33612, United States
| | - Tina S. Mutka
- Department
of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, Florida 33612, United States
| | - Ala Azhari
- Department
of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, Florida 33612, United States
| | - Dennis E. Kyle
- Department
of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, Florida 33612, United States
| | - James W. Leahy
- Department of Chemistry, University of South Florida, CHE 205, 4202 East Fowler Avenue, Tampa, Florida 33620, United States
- Florida Center of Excellence for Drug Discovery
and Innovation, University of South Florida, 3720 Spectrum Boulevard, Suite 303, Tampa, Florida 33612, United States
- Department of
Molecular Medicine, Morsani College of Medicine, University of South Florida, MDC 7, 12901 Bruce B. Downs Boulevard, Tampa, Florida 33612, United States
| |
Collapse
|
19
|
Murillo-Solano C, Dong C, Sanchez CG, Pizarro JC. Identification and characterization of the antiplasmodial activity of Hsp90 inhibitors. Malar J 2017; 16:292. [PMID: 28724415 PMCID: PMC5518105 DOI: 10.1186/s12936-017-1940-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/14/2017] [Indexed: 01/13/2023] Open
Abstract
Background The recent reduction in mortality due to malaria is being threatened by the appearance of Plasmodium falciparum parasites that are resistant to artemisinin in Southeast Asia. To limit the impact of resistant parasites and their spread across the world, there is a need to validate anti-malarial drug targets and identify new leads that will serve as foundations for future drug development programmes targeting malaria. Towards that end, the antiplasmodial potential of several Hsp90 inhibitors was characterized. Because, the Hsp90 chaperone has been suggested as a good drug target against multiple parasitic infections including malaria. Results Chemically diverse sets of Hsp90 inhibitors, evaluated in clinical trials as anti-cancer agents, were tested against the malaria parasite. Most of the compounds showed strong antiplasmodial activity in growth inhibition assays against chloroquine sensitive and resistant strains. There was a good agreement between the compound in vitro anti-parasitic activity and their affinity against the Plasmodium chaperone. The two most potent Hsp90 inhibitors also showed cytocidal activity against two P. falciparum strains. Their antiplasmodial activity affected all parasite forms during the malaria blood cycle. However, the compounds activity against the parasite showed no synergy when combined with anti-malarial drugs, like chloroquine or DHA. Discussion The Hsp90 inhibitors anti-parasitic activity correlates with their affinity to their predicted target the P. falciparum chaperone Hsp90. However, the most effective compounds also showed high affinity for a close homologue, Grp94. This association points to a mode of action for Hsp90 inhibitors that correlate compound efficacy with multi-target engagement. Besides their ability to limit parasite replication, two compounds also significantly impacted P. falciparum viability in vitro. Finally, a structural analysis suggests that the best hit represents a promising scaffold to develop parasite specific leads according. Conclusion The results shown that Hsp90 inhibitors are lethal against the malaria parasite. The correlation between biochemical and in vitro data strongly supports Hsp90 as a drug target against the malaria parasite. Furthermore, at least one Hsp90 inhibitor developed as anticancer therapeutics could serve as starting point to generate P. falciparum-specific lead compounds. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1940-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claribel Murillo-Solano
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Chunmin Dong
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Cecilia G Sanchez
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA. .,Vector-Borne Infectious Diseases Research Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
20
|
Bayih AG, Folefoc A, Mohon AN, Eagon S, Anderson M, Pillai DR. In vitro and in vivo anti-malarial activity of novel harmine-analog heat shock protein 90 inhibitors: a possible partner for artemisinin. Malar J 2016; 15:579. [PMID: 27903279 PMCID: PMC5131496 DOI: 10.1186/s12936-016-1625-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/18/2016] [Indexed: 11/13/2022] Open
Abstract
Background The emergence of artemisinin-resistant Plasmodium falciparum strains poses a serious challenge to the control of malaria. This necessitates the development of new anti-malarial drugs. Previous studies have shown that the natural beta-carboline alkaloid harmine is a promising anti-malarial agent targeting the P. falciparum heat-shock protein 90 (PfHsp90). The aim of this study was to test the anti-malarial activity of harmine analogues. Methods Forty-two harmine analogues were synthesized and the binding of these analogues to P. falciparum heat shock protein 90 was investigated. The in vitro anti-malarial activity of two of the analogues, 17A and 21A, was evaluated using a 72-h growth inhibition assay. The in vivo anti-malarial activity was tested in Plasmodium berghei infection of BALB/c mice. The potential of 21A for a combination treatment with artemisinin was evaluated using in vivo combination study with dihydro-artemisinin in BALB/c mice. Cytotoxicity of the harmine analogues was tested in vitro using HepG2 and HeLa cell lines. Results 17A and 21A bound to PfHsp90 with average IC50 values of 12.2 ± 2.3 and 23.1 ± 8.8 µM, respectively. They also inhibited the P. falciparum W2 strain with average IC50 values of 4.2 ± 1.3 and 5.7 ± 1.7 µM, respectively. In vivo, three daily injections of P. berghei-infected BALB/c mice with 100 mg/kg of either 17A or 21A showed significant reduction in parasitaemia with a 51.5 and 56.1% reduction, respectively. Mice treated with 17A and 21A showed a median survival time of 11 and 14 days, respectively, while the vehicle control mice survived a median of only 8.5 days. A dose-ranging experiment with 21A showed that the compound has a dose-dependent anti-malarial effect. Furthermore, treatment of infected mice with a combination of 21A and dihydroartemisinin (DHA) showed a dramatic reduction in parasitaemia compared to treatment with DHA alone. Conclusion A novel and non-toxic harmine analogue has been synthesized which binds to PfHsp90 protein, inhibits P. falciparum in vitro at micromolar concentration, reduces parasitaemia and prolongs survival of P. berghei-infected mice with an additive anti-malarial effect when combined with DHA.
Collapse
Affiliation(s)
- Abebe Genetu Bayih
- Department of Pathology and Laboratory Medicine, MIID and Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Medical Parasitology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Asongna Folefoc
- Department of Pathology and Laboratory Medicine, MIID and Medicine, University of Calgary, Calgary, AB, Canada
| | - Abu Naser Mohon
- Department of Pathology and Laboratory Medicine, MIID and Medicine, University of Calgary, Calgary, AB, Canada
| | - Scott Eagon
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Marc Anderson
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, CA, USA
| | - Dylan R Pillai
- Department of Pathology and Laboratory Medicine, MIID and Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
Yamasaki M, Tsuboi Y, Taniyama Y, Uchida N, Sato R, Nakamura K, Ohta H, Takiguchi M. Molecular cloning, phylogenetic analysis and heat shock response of Babesia gibsoni heat shock protein 90. J Vet Med Sci 2016; 78:1355-60. [PMID: 27149891 PMCID: PMC5053942 DOI: 10.1292/jvms.16-0027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Babesia gibsoni heat shock protein 90 (BgHSP90)
gene was cloned and sequenced. The length of the gene was 2,610 bp with two introns. This
gene was amplified from cDNA corresponding to full length coding sequence (CDS) with an
open reading frame of 2,148 bp. A phylogenetic analysis of the CDS of
HSP90 gene showed that B. gibsoni was most closely
related to B. bovis and Babesia sp. BQ1/Lintan and lies
within a phylogenetic cluster of protozoa. Moreover, mRNA transcription profile for
BgHSP90 exposed to high temperature were examined by quantitative
real-time reverse transcription-polymerase chain reaction. BgHSP90 levels
were elevated when the parasites were incubated at 43°C for 1 hr.
Collapse
Affiliation(s)
- Masahiro Yamasaki
- Laboratory of Veterinary Small Animal Internal Medicine, Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Giannini G, Battistuzzi G. Exploring in vitro and in vivo Hsp90 inhibitors activity against human protozoan parasites. Bioorg Med Chem Lett 2014; 25:462-5. [PMID: 25547934 DOI: 10.1016/j.bmcl.2014.12.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 11/20/2022]
Abstract
A set of compounds, previously selected as potent Hsp90α inhibitors, has been studied on a panel of human parasites. 5-Aryl-3,4-isoxazolediamide derivatives (1) were active against two protozoa, Trypanosoma brucei rhodesiense and Plasmodium falciparum, with a good tolerability toward cytotoxicity on non-malignant L6 rat myoblast cell line, unlike the 1,5-diaryl,4-carboxamides-1,2,3-triazole derivatives (2) which, while showing a single-digit nM range activity against the same protozoa, were also highly cytotoxic on L6 cells. In a subsequent in vivo study, two isoxazolediamide derivatives, 1a and 1b, were very efficacious on the sleeping sickness-causing agent with a clear parasitaemia during treatment. These data, however, showed that not all protozoa are sensitive to Hsp90 inhibitors, as well as not all Hsp90 inhibitors are equally active on parasites.
Collapse
Affiliation(s)
- Giuseppe Giannini
- R&D Sigma-Tau Industrie Farmaceutiche Riunite S.p.A., Via Pontina Km 30,400, I-00040, Pomezia, Roma, Italy.
| | - Gianfranco Battistuzzi
- R&D Sigma-Tau Industrie Farmaceutiche Riunite S.p.A., Via Pontina Km 30,400, I-00040, Pomezia, Roma, Italy
| |
Collapse
|
23
|
Khan MK, He L, Zhang W, Wang Y, Tao Q, Song Q, Sajid MS, Yu Q, Hu J, Fang R, Hu M, Zhou Y, Zhao J. Identification of two novel HSP90 proteins in Babesia orientalis: molecular characterization, and computational analyses of their structure, function, antigenicity and inhibitor interaction. Parasit Vectors 2014; 7:293. [PMID: 24970594 PMCID: PMC4089566 DOI: 10.1186/1756-3305-7-293] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 06/17/2014] [Indexed: 11/13/2022] Open
Abstract
Background HSP90 protects the cells from heat stress and facilitates protein maturation and stability. The full genome sequences of piroplasms contain two putative HSP90 proteins, which are yet uncharacterized. To this end, the two putative HSP90 proteins of Babesia orientalis were identified and characterized by molecular and in silico methods. Methods The two putative proteins in B. orientalis genome showing homology with putative HSP90 of other piroplasms were cloned and sequenced. A computational analysis was carried out to predict the antigenic determinants, structure and function of these proteins. The interactions of two HSP90 isoforms with respective inhibitors were also examined through docking analysis. Results The length of BoHSP90-A gene (amplified from gDNA) was 2706 bp with one intron from position 997 to 1299 bp. This gene amplified from cDNA corresponded to full length CDS with an open reading frame (ORF) of 2403 bp encoding a 800 amino acid (AA) polypeptide with a predicted size of 91.02 kDa. The HSP90-B gene was intronless with an ORF of 2349 bp, and predicted polypeptide comprised of 797 AA with a size of 90.59 kDa. The AA sequences of these two proteins of B. orientalis were the most identical to those of B. bovis. The BoHSP90-A and BoHSP90-B were recognized as 90 kDa in the parasite lysate by the rabbit antisera raised against the recombinant BoHSP90 proteins. The anti-B. orientalis buffalo serum reacted with the rBoHSP90s expressed in E. coli, indicating that these proteins might be secreted by the parasite before entry into host cells. The overall structure and functional analyses showed several domains involved in ATPase activity, client protein binding and HSP90 dimerization. Likewise, several HSP90 inhibitors showed binding to ATP binding pockets of BoHSP90-A and BoHSP90-B, as observed through protein structure-ligand interaction analysis. Conclusions The two putative HSP90 proteins in B. orientalis were recognized as 90 kDa. The rBoHSP90-A and rBoHSP90-B were reacted with the B. orientalis infected buffalo serum. The computational structure and functional analyses revealed that these two proteins may have chaperonic activity. The protein structure-ligand interaction analyses indicated that these two proteins had many drug target sites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
24
|
Abstract
Trypanosomiasis is caused by Trypanosoma species which affect both human and animal populations and pose a major threat to developing countries. The incidence of animal trypanosomiasis is on the rise. Surra is a type of animal trypanosomiasis, caused by Trypanosoma evansi, and has been included in priority list B of significant diseases by the World Organization of Animal Health (OIE). Control of surra has been a challenge due to the lack of effective drugs and vaccines and emergence of resistance towards existing drugs. Our laboratory has previously implicated Heat shock protein 90 (Hsp90) from protozoan parasites as a potential drug target and successfully demonstrated efficacy of an Hsp90 inhibitor in cell culture as well as a pre-clinical mouse model of trypanosomiasis. This article explores the role of Hsp90 in the Trypanosoma life cycle and its potential as a drug target. It appears plausible that the repertoire of Hsp90 inhibitors available in academia and industry may have value for treatment of surra and other animal trypanosomiasis.
Collapse
|
25
|
Nageshan RK, Roy N, Ranade S, Tatu U. Trans-spliced heat shock protein 90 modulates encystation in Giardia lamblia. PLoS Negl Trop Dis 2014; 8:e2829. [PMID: 24786776 PMCID: PMC4006730 DOI: 10.1371/journal.pntd.0002829] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/13/2014] [Indexed: 01/22/2023] Open
Abstract
Background Hsp90 from Giardia lamblia is expressed by splicing of two independently transcribed RNA molecules, coded by genes named HspN and HspC located 777 kb apart. The reasons underlying such unique trans-splicing based generation of GlHsp90 remain unclear. Principle Finding In this study using mass-spectrometry we identify the sequence of the unique, junctional peptide contributed by the 5′ UTR of HspC ORF. This peptide is critical for the catalytic function of Hsp90 as it harbours an essential “Arg” in its sequence. We also show that full length GlHsp90 possesses all the functional hall marks of a canonical Hsp90 including its ability to bind and hydrolyze ATP. Using qRT-PCR as well as western blotting approach we find the reconstructed Hsp90 to be induced in response to heat shock. On the contrary we find GlHsp90 to be down regulated during transition from proliferative trophozoites to environmentally resistant cysts. This down regulation of GlHsp90 appears to be mechanistically linked to the encystation process as we find pharmacological inhibition of GlHsp90 function to specifically induce encystation. Significance Our results implicate the trans-spliced GlHsp90 from Giardia lamblia to regulate an essential stage transition in the life cycle of this important human parasite. Giardia lamblia is one of the most common causes of diarrhoea across the globe. The disease can result in fatalities especially in small children. The parasite is transmitted by contaminated food through faeco-oral route due to unhygienic habits. The parasite exhibits two stages during its lifecycle; namely cysts and trophozoites. Due to their environmentally resistant hardy nature cysts are transmitted through contaminated food into the human body. Upon entry into the human body they convert into active trophozoites and cause pathogenesis of the disease. In the course of infection within the host, some of the trophozoites convert back into cysts and are released in the environment through the faeces. The mechanisms and signals that convert the parasite from trophozoites to cysts are not yet known. Our study, for the first time, implicates heat shock protein 90 of the parasite in the conversion of trophozoites into cysts in the intestine of the infected human body. Hsp90 is famous for its ability to sense environmental changes and provide cues for stage-switch in related parasites. In addition to providing a glimpse into molecular mechanisms of stage inter-conversion, our results suggest potential new ways of treating this important human infection.
Collapse
Affiliation(s)
| | - Nainita Roy
- The Department of Biochemistry, Indian Institute of Sciences, Bangalore, India
| | - Shatakshi Ranade
- The Department of Biochemistry, Indian Institute of Sciences, Bangalore, India
| | - Utpal Tatu
- The Department of Biochemistry, Indian Institute of Sciences, Bangalore, India
- * E-mail:
| |
Collapse
|
26
|
Wang T, Bisson WH, Mäser P, Scapozza L, Picard D. Differences in conformational dynamics between Plasmodium falciparum and human Hsp90 orthologues enable the structure-based discovery of pathogen-selective inhibitors. J Med Chem 2014; 57:2524-35. [PMID: 24580531 DOI: 10.1021/jm401801t] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The high sequence conservation of druggable pockets of closely related proteins can make it challenging to develop selective inhibitors. We designed a new drug discovery approach that exploits both the static and dynamic differences of two orthologues. We applied it, as a proof of concept, to identify compounds that discriminate between the molecular chaperone Hsp90 of the protozoan pathogen Plasmodium falciparum (Pf) and that of its human host. We found that the ATP-binding pocket has a Pf-specific extension, whose sequence lining is identical in human Hsp90 but which differs by tertiary structure and dynamics. Using these insights for a structure-based drug screen, we discovered novel 7-azaindole compounds that exclusively bind the recombinant N-terminal domain of PfHsp90 but not of human Hsp90 nor of a PfHsp90 mutant with "human-like" dynamics. Moreover, these compounds preferentially inhibit the growth of yeast complemented by PfHsp90 and block the growth of Pf in culture.
Collapse
Affiliation(s)
- Tai Wang
- Department of Cell Biology, University of Geneva , and ‡Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences of the Universities of Geneva and Lausanne , 30 Quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland
| | | | | | | | | |
Collapse
|
27
|
Zettler J, Xia H, Burkard N, Kulik A, Grond S, Heide L, Apel AK. New aminocoumarins from the rare actinomycete Catenulispora acidiphila DSM 44928: identification, structure elucidation, and heterologous production. Chembiochem 2014; 15:612-21. [PMID: 24554531 DOI: 10.1002/cbic.201300712] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Indexed: 11/08/2022]
Abstract
Genome mining led to the discovery of a novel aminocoumarin gene cluster in the rare actinomycete Catenulispora acidiphila DSM 44928. Sequence analysis revealed the presence of genes putatively involved in export/resistance, regulation, and biosynthesis of the aminocoumarin moiety and its halogenation, as well as several genes with so far unknown function. Two new aminocoumarins, cacibiocin A and B, were identified in the culture broth of C. acidiphila. Heterologous expression of the putative gene cluster in Streptomyces coelicolor M1152 confirmed that this cluster is responsible for cacibiocin biosynthesis. Furthermore, total production levels of cacibiocins could be increased by heterologous expression and screening of different culture media from an initial yield of 4.9 mg L(-1) in C. acidiphila to 60 mg L(-1) in S. coelicolor M1152. By HR-MS and NMR analysis, cacibiocin A was found to contain a 3-amino-4,7-dihydroxycoumarin moiety linked by an amide bond to a pyrrole-2,5-dicarboxylic acid. The latter structural motif has not been identified previously in any natural compound. Additionally, cacibiocin B contains two chlorine atoms at positions 6' and 8' of the aminocoumarin moiety.
Collapse
Affiliation(s)
- Judith Zettler
- Eberhard-Karls-Universität Tübingen, Pharmazeutische Biologie, Auf der Morgenstelle 8, 72076 Tübingen (Germany); German Centre for Infection Research (DZIF), Partner site Tübingen (Germany)
| | | | | | | | | | | | | |
Collapse
|
28
|
Pizarro JC, Hills T, Senisterra G, Wernimont AK, Mackenzie C, Norcross NR, Ferguson MAJ, Wyatt PG, Gilbert IH, Hui R. Exploring the Trypanosoma brucei Hsp83 potential as a target for structure guided drug design. PLoS Negl Trop Dis 2013; 7:e2492. [PMID: 24147171 PMCID: PMC3798429 DOI: 10.1371/journal.pntd.0002492] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 09/09/2013] [Indexed: 01/08/2023] Open
Abstract
Human African trypanosomiasis is a neglected parasitic disease that is fatal if untreated. The current drugs available to eliminate the causative agent Trypanosoma brucei have multiple liabilities, including toxicity, increasing problems due to treatment failure and limited efficacy. There are two approaches to discover novel antimicrobial drugs - whole-cell screening and target-based discovery. In the latter case, there is a need to identify and validate novel drug targets in Trypanosoma parasites. The heat shock proteins (Hsp), while best known as cancer targets with a number of drug candidates in clinical development, are a family of emerging targets for infectious diseases. In this paper, we report the exploration of T. brucei Hsp83 – a homolog of human Hsp90 – as a drug target using multiple biophysical and biochemical techniques. Our approach included the characterization of the chemical sensitivity of the parasitic chaperone against a library of known Hsp90 inhibitors by means of differential scanning fluorimetry (DSF). Several compounds identified by this screening procedure were further studied using isothermal titration calorimetry (ITC) and X-ray crystallography, as well as tested in parasite growth inhibitions assays. These experiments led us to the identification of a benzamide derivative compound capable of interacting with TbHsp83 more strongly than with its human homologs and structural rationalization of this selectivity. The results highlight the opportunities created by subtle structural differences to develop new series of compounds to selectively target the Trypanosoma brucei chaperone and effectively kill the sleeping sickness parasite. Sleeping sickness, or human African trypanosomiasis (HAT), is a deadly neglected disease for which new therapeutic options are badly needed. Current drugs have several liabilities including toxicity and route of administration limiting their efficacy to combat the disease. Our study aimed at validating a potential new drug target against Trypanosoma brucei, its heat shock protein 83 (Hsp83). The chaperone was screened against a repurposed library composed of inhibitors against the human Hsp90. The compounds were assayed in their ability to bind the T. brucei protein and to kill the parasite. Our work has identified selective and high-affinity chemical compounds targeting the parasitic Hsp83. Additionally, structural studies were conducted to explore the observed selectivity of selected inhibitors. Our work has validated T. brucei Hsp83 as a potential target for future drug discovery campaigns. It has also shown the strength of repurposing chemical libraries developed against human proteins, emphasizing the possibility to piggyback current and past drug discovery efforts for other diseases in the search for new drugs against neglected tropical diseases.
Collapse
Affiliation(s)
- Juan Carlos Pizarro
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| | - Tanya Hills
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Guillermo Senisterra
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Amy K. Wernimont
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Claire Mackenzie
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Neil R. Norcross
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Michael A. J. Ferguson
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Paul G. Wyatt
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Ian H. Gilbert
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Raymond Hui
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|