1
|
Ortega-Caballero M, Gonzalez-Vazquez MC, Hernández-Espinosa MA, Carabarin-Lima A, Mendez-Albores A. The Impact of Environmental and Housing Factors on the Distribution of Triatominae (Hemiptera, Reduviidae) in an Endemic Area of Chagas Disease in Puebla, Mexico. Diseases 2024; 12:238. [PMID: 39452481 PMCID: PMC11506842 DOI: 10.3390/diseases12100238] [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: 07/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Chagas disease (CD), a Neglected Tropical Disease caused by Trypanosoma cruzi, affects millions of people in Latin America and the southern US and spreads worldwide. CD results from close interactions between humans, animals, and vectors, influenced by sociodemographic factors and housing materials. METHODS This study aimed to evaluate how these factors, along with seasonal changes, affect the distribution of CD vectors in an endemic community near Puebla, Mexico, using a cross-sectional survey. A total of 383 people from this area, known for the presence of major vectors such as Triatoma barberi and Triatoma pallidipennis, were surveyed. RESULTS As a result of the survey, it was found that only 27.4% of respondents knew about CD, and 83.3% owned potential reservoir pets; additionally, the quality of the wall, roof, and floor significantly influenced vector sightings, while the seasonal pattern showed less of an association. Chi-square tests confirmed these associations between vector sightings and housing materials (p < 0.001); vector sightings versus seasonal patterns showed less of an association (p = 0.04), and land use changes did not show an association (p = 0.27). CONCLUSIONS Construction materials play an important role in the sighting of triatomines in homes, so important actions should be taken to improve homes. However, further experimental or longitudinal studies are needed to establish causality.
Collapse
Affiliation(s)
- Miguel Ortega-Caballero
- Centro de Química-ICUAP-Posgrado en Ciencias Ambientales, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (M.O.-C.); (M.A.H.-E.)
| | | | - Miguel Angel Hernández-Espinosa
- Centro de Química-ICUAP-Posgrado en Ciencias Ambientales, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (M.O.-C.); (M.A.H.-E.)
| | - Alejandro Carabarin-Lima
- Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico
| | - Alia Mendez-Albores
- Centro de Química-ICUAP-Posgrado en Ciencias Ambientales, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (M.O.-C.); (M.A.H.-E.)
| |
Collapse
|
2
|
Tristão DC, Barbosa H, de Castro Levatti EV, Andrade BA, Romanelli MM, Antar GM, Tempone AG, Lago JHG. Selective Activity Against Amastigote Forms of Trypanosoma cruzi and Leishmania infantum of Diastereomeric Dicentrine N-oxides. Chem Biodivers 2024; 21:e202401247. [PMID: 38896778 DOI: 10.1002/cbdv.202401247] [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: 05/16/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/21/2024]
Abstract
As part of our continuous research for the discovery of bioactive compounds against Trypanosoma cruzi and Leishmania infantum, the alkaloid (6aS)-dicentrine (1) was oxidized to afford (6aS,6S)- (2) and (6aS,6R)- (3) dicentrine-N-oxides. Evaluation of the cytotoxicity against NCTC cells indicated that 2 and 3 are non-toxic (CC50>200 μM) whereas 1 demonstrated CC50 of 52.0 μM. Concerning T. cruzi activity against amastigotes, derivatives 2 and 3 exhibited EC50 values of 9.9 μM (SI>20.2) and 27.5 μM (SI>7.3), respectively, but 1 is inactive (EC50>100 μM). Otherwise, when tested against L. infantum amastigotes, 1 and 3 exhibited EC50 values of 10.3 μM (SI=5.0) and 12.7 μM (SI>15.7), respectively, being 2 inactive (EC50>100 μM). Comparing the effects of positive controls benznidazol (EC50=6.5 μM and SI>30.7) and miltefosine (EC50=10.2 μM and SI=15.2), it was observed a selective antiparasitic activity to diastereomers 2 and 3 against T. cruzi and L. infantum. Considering stereochemical aspects, it was suggested that the configuration of the new stereocenter formed after oxidation of 1 played an important role in the bioactivity against amastigotes of both tested parasites.
Collapse
Affiliation(s)
- Daniela C Tristão
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | - Henrique Barbosa
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | | | | | | | - Guilherme M Antar
- Department of Agricultural and Biological Sciences, Federal University of Espirito Santo, 29932-540, São Matheus, ES, Brazil
| | | | - João Henrique G Lago
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| |
Collapse
|
3
|
Rana MM, Lu S, Menjívar TA, Fukaya K, Nakajima-Shimada J, Urabe D, Igarashi Y. Cladoic Acid, an Anti- Trypanosoma cruzi Polyketide from Cladosporium sp. TP-F2020. JOURNAL OF NATURAL PRODUCTS 2024; 87:2126-2131. [PMID: 39101838 DOI: 10.1021/acs.jnatprod.4c00567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
A new polyketide, cladoic acid, was isolated from a fungus of the genus Cladosporium. The structure of the highly oxygenated trans-decalin ring with an all-E triene side chain was elucidated by extensive spectroscopic analysis. The unique chair/twist-boat conformation of the trans-decalin core and the flexibility of the B-ring were demonstrated by computer-aided conformational analysis. Cladoic acid was active against Trypanosoma cruzi and inhibited the proliferation of amastigotes and epimastigotes with IC50 values of 27 and 46 μM, respectively, but it did not show any appreciable activity against P388 murine leukemia cells, bacteria, or fungi, indicating it is a potential candidate for drug development against Chagas disease.
Collapse
Affiliation(s)
- Md Masud Rana
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Shiyang Lu
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Tatiana Ascencio Menjívar
- Department of Microbiology, School of Medicine, Dr. José Matías Delgado University, Campus 1, km 8 1/2 Carretera a Santa Tecla, San Salvador 01101, El Salvador
- Graduate School of Health Sciences, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8510, Japan
| | - Keisuke Fukaya
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Junko Nakajima-Shimada
- Graduate School of Health Sciences, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8510, Japan
| | - Daisuke Urabe
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| |
Collapse
|
4
|
Gangwar U, Choudhury H, Shameem R, Singh Y, Bansal A. Recent development in CRISPR-Cas systems for human protozoan diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 208:109-160. [PMID: 39266180 DOI: 10.1016/bs.pmbts.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Protozoan parasitic diseases pose a substantial global health burden. Understanding the pathogenesis of these diseases is crucial for developing intervention strategies in the form of vaccine and drugs. Manipulating the parasite's genome is essential for gaining insights into its fundamental biology. Traditional genomic manipulation methods rely on stochastic homologous recombination events, which necessitates months of maintaining the cultured parasites under drug pressure to generate desired transgenics. The introduction of mega-nucleases (MNs), zinc-finger nucleases (ZFNs), and transcription activator-like effector nucleases (TALENs) greatly reduced the time required for obtaining a desired modification. However, there is a complexity associated with the design of these nucleases. CRISPR (Clustered regularly interspaced short palindromic repeats)/Cas (CRISPR associated proteins) is the latest gene editing tool that provides an efficient and convenient method for precise genomic manipulations in protozoan parasites. In this chapter, we have elaborated various strategies that have been adopted for the use of CRISPR-Cas9 system in Plasmodium, Leishmania and Trypanosoma. We have also discussed various applications of CRISPR-Cas9 pertaining to understanding of the parasite biology, development of drug resistance mechanism, gene drive and diagnosis of the infection.
Collapse
Affiliation(s)
- Utkarsh Gangwar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Risha Shameem
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Yashi Singh
- Department of Biosciences & Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Abhisheka Bansal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
5
|
Rodríguez-Garza NE, Quintanilla-Licea R, Gomez-Flores R, Galaviz-Silva L, Molina-Garza ZJ. In Vitro Antiprotozoal Activity of Schinus molle Extract, Partitions, and Fractions against Trypanosoma cruzi. PLANTS (BASEL, SWITZERLAND) 2024; 13:2177. [PMID: 39204613 PMCID: PMC11359525 DOI: 10.3390/plants13162177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, represents an important and worldwide public health issue, particularly in Latin America. Limitations of conventional treatment with benznidazole and nifurtimox underscore the urgent need for new therapeutic strategies for this disease. Schinus molle, a tree used in traditional medicine for various ailments, has demonstrated promising antiparasitic activity. The in vitro anti-T. cruzi activity of Schinus molle crude methanol extract, partitions, and fractions, as well as their cytotoxicity in Vero cells and Artemia salina, and hemolytic activity in human erythrocytes were assessed. Most of the extracts possessed anti-T. cruzi effects, with Sm-CF3 being the fraction with the highest activity (IC50 = 19 µg/mL; SI = 6.8). Gas chromatography-mass spectrometry analysis identified 20 compounds, with fatty acyls comprising the predominant chemical class (55%). We also identified the antiparasitic compounds cis-5,8,11,14,17-eicosapentaenoic acid and trans-Z-α-bisabolene epoxide, suggesting their potential contribution to the observed anti-T. cruzi activity. In conclusion, our findings support the therapeutic potential of S. molle as a source of novel antiparasitic agents against T. cruzi.
Collapse
Affiliation(s)
- Nancy E Rodríguez-Garza
- Laboratorio de Patología Molecular y Experimental, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza 66455, Nuevo León, Mexico
| | - Ramiro Quintanilla-Licea
- Laboratorio de Fitoquímica, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza 66455, Nuevo León, Mexico
| | - Ricardo Gomez-Flores
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza 66455, Nuevo León, Mexico
| | - Lucio Galaviz-Silva
- Laboratorio de Patología Molecular y Experimental, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza 66455, Nuevo León, Mexico
| | - Zinnia J Molina-Garza
- Laboratorio de Patología Molecular y Experimental, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza 66455, Nuevo León, Mexico
| |
Collapse
|
6
|
Vázquez ME, Zabala BA, Mesías AC, Biscari L, Kaufman CD, Alloatti A, Siano F, Picariello G, Corbalán NS, Lenis BA, Toscano MA, Parodi CM, Brandán CMP, Acuña L. Protective Efficacy of the Epitope-Conjugated Antigen N-Tc52/TSkb20 in Mitigating Trypanosoma cruzi Infection through CD8+ T-Cells and IFNγ Responses. Vaccines (Basel) 2024; 12:621. [PMID: 38932350 PMCID: PMC11209121 DOI: 10.3390/vaccines12060621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, remains a major public health challenge affecting millions in Latin America and worldwide. Although significant progress has been made in vector control, no vaccine exists to prevent infection or mitigate disease pathogenesis. We developed a rationally designed chimeric protein vaccine, N-Tc52/TSkb20, incorporating immunodominant epitopes from two T. cruzi antigens, the amino-terminal portion of Tc52 and the TSkb20 epitope derived from trans-sialidase. The objectives of this study were to construct and characterize the antigen and evaluate its protective potential in an immunoprophylactic murine model of T. cruzi infection. The N-Tc52/TSkb20 protein was recombinantly expressed in E. coli and its identity was confirmed using mass spectrometry and Western blotting. Immunization with the chimeric protein significantly controlled parasitemia and reduced the heart, colon, and skeletal muscle parasite burdens compared to non-vaccinated mice. Protection was superior to vaccination with the individual parental antigen components. Mechanistically, the vaccine induced potent CD8+ T-cell and IFNγ responses against the incorporated epitopes and a protective IgG antibody profile. A relatively low IL-10 response favored early parasite control. These results validate the promising multi-epitope approach and support the continued development of this type of rational vaccine design strategy against Chagas disease.
Collapse
Affiliation(s)
- María Elisa Vázquez
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Brenda A. Zabala
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Andrea C. Mesías
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Lucia Biscari
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Cintia D. Kaufman
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Andrés Alloatti
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Francesco Siano
- Istituto di Scienze dell’ Alimentazione—Consiglio Nazionale delle Ricerche (CNR), 83100 Avellino, Italy; (F.S.); (G.P.)
| | - Gianluca Picariello
- Istituto di Scienze dell’ Alimentazione—Consiglio Nazionale delle Ricerche (CNR), 83100 Avellino, Italy; (F.S.); (G.P.)
| | - Natalia S. Corbalán
- Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta A4400, Argentina;
| | - Bladimiro A. Lenis
- Unidad de Conocimiento Traslacional, Hospital Arturo Oñativia, Salta A4400, Argentina; (B.A.L.); (M.A.T.)
| | - Marta A. Toscano
- Unidad de Conocimiento Traslacional, Hospital Arturo Oñativia, Salta A4400, Argentina; (B.A.L.); (M.A.T.)
| | - Cecilia M. Parodi
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Cecilia M. Pérez Brandán
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Leonardo Acuña
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| |
Collapse
|
7
|
Trajano-Silva LAM, Mule SN, Palmisano G. Molecular tools to regulate gene expression in Trypanosoma cruzi. Adv Clin Chem 2024; 120:169-190. [PMID: 38762241 DOI: 10.1016/bs.acc.2024.04.008] [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] [Indexed: 05/20/2024]
Abstract
Developing molecular strategies to manipulate gene expression in trypanosomatids is challenging, particularly with respect to the unique gene expression mechanisms adopted by these unicellular parasites, such as polycistronic mRNA transcription and multi-gene families. In the case of Trypanosoma cruzi (T. cruzi), the causative agent of Chagas Disease, the lack of RNA interference machinery further complicated functional genetic studies important for understanding parasitic biology and developing biomarkers and potential therapeutic targets. Therefore, alternative methods of performing knockout and/or endogenous labelling experiments were developed to identify and understand the function of proteins for survival and interaction with the host. In this review, we present the main tools for the genetic manipulation of T. cruzi, focusing on the Clustered Regularly Interspaced Short Palindromic Repeats Cas9-associated system technique widely used in this organism. Moreover, we highlight the importance of using these tools to elucidate the function of uncharacterized and glycosylated proteins. Further developments of these technologies will allow the identification of new biomarkers, therapeutic targets and potential vaccines against Chagas disease with greater efficiency and speed.
Collapse
Affiliation(s)
- Lays Adrianne M Trajano-Silva
- Glycoproteomic Laboratory, Parasitology Department, Institute of Biomedical Science II, University of Sao Paulo, Sao Paulo, Brazil
| | - Simon Ngao Mule
- Glycoproteomic Laboratory, Parasitology Department, Institute of Biomedical Science II, University of Sao Paulo, Sao Paulo, Brazil
| | - Giuseppe Palmisano
- Glycoproteomic Laboratory, Parasitology Department, Institute of Biomedical Science II, University of Sao Paulo, Sao Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Stadler KA, Ortiz-Joya LJ, Singh Sahrawat A, Buhlheller C, Gruber K, Pavkov-Keller T, O'Hagan TB, Guarné A, Pulido S, Marín-Villa M, Zangger K, Gubensäk N. Structural investigation of Trypanosoma cruzi Akt-like kinase as drug target against Chagas disease. Sci Rep 2024; 14:10039. [PMID: 38693166 PMCID: PMC11063076 DOI: 10.1038/s41598-024-59654-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
According to the World Health Organization, Chagas disease (CD) is the most prevalent poverty-promoting neglected tropical disease. Alarmingly, climate change is accelerating the geographical spreading of CD causative parasite, Trypanosoma cruzi, which additionally increases infection rates. Still, CD treatment remains challenging due to a lack of safe and efficient drugs. In this work, we analyze the viability of T. cruzi Akt-like kinase (TcAkt) as drug target against CD including primary structural and functional information about a parasitic Akt protein. Nuclear Magnetic Resonance derived information in combination with Molecular Dynamics simulations offer detailed insights into structural properties of the pleckstrin homology (PH) domain of TcAkt and its binding to phosphatidylinositol phosphate ligands (PIP). Experimental data combined with Alpha Fold proposes a model for the mechanism of action of TcAkt involving a PIP-induced disruption of the intramolecular interface between the kinase and the PH domain resulting in an open conformation enabling TcAkt kinase activity. Further docking experiments reveal that TcAkt is recognized by human inhibitors PIT-1 and capivasertib, and TcAkt inhibition by UBMC-4 and UBMC-6 is achieved via binding to TcAkt kinase domain. Our in-depth structural analysis of TcAkt reveals potential sites for drug development against CD, located at activity essential regions.
Collapse
Affiliation(s)
- Karina A Stadler
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria
| | - Lesly J Ortiz-Joya
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Amit Singh Sahrawat
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Innophore GmbH, Graz, Austria
| | | | - Karl Gruber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Innophore GmbH, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | | - Alba Guarné
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Sergio Pulido
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- LifeFactors ZF SAS, Rionegro, Colombia
| | - Marcel Marín-Villa
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Klaus Zangger
- Institute of Chemistry/Organic and Bioorganic Chemistry, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Nina Gubensäk
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| |
Collapse
|
9
|
Huffman A, Zhang X, Lanka M, Zheng J, Masci AM, He Y. Ontological representation, modeling, and analysis of parasite vaccines. J Biomed Semantics 2024; 15:4. [PMID: 38664818 PMCID: PMC11044459 DOI: 10.1186/s13326-024-00307-0] [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: 11/01/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Pathogenic parasites are responsible for multiple diseases, such as malaria and Chagas disease, in humans and livestock. Traditionally, pathogenic parasites have been largely an evasive topic for vaccine design, with most successful vaccines only emerging recently. To aid vaccine design, the VIOLIN vaccine knowledgebase has collected vaccines from all sources to serve as a comprehensive vaccine knowledgebase. VIOLIN utilizes the Vaccine Ontology (VO) to standardize the modeling of vaccine data. VO did not model complex life cycles as seen in parasites. With the inclusion of successful parasite vaccines, an update in parasite vaccine modeling was needed. RESULTS VIOLIN was expanded to include 258 parasite vaccines against 23 protozoan species, and 607 new parasite vaccine-related terms were added to VO since 2022. The updated VO design for parasite vaccines accounts for parasite life stages and for transmission-blocking vaccines. A total of 356 terms from the Ontology of Parasite Lifecycle (OPL) were imported to VO to help represent the effect of different parasite life stages. A new VO class term, 'transmission-blocking vaccine,' was added to represent vaccines able to block infectious transmission, and one new VO object property, 'blocks transmission of pathogen via vaccine,' was added to link vaccine and pathogen in which the vaccine blocks the transmission of the pathogen. Additionally, our Gene Set Enrichment Analysis (GSEA) of 140 parasite antigens used in the parasitic vaccines identified enriched features. For example, significant patterns, such as signal, plasma membrane, and entry into host, were found in the antigens of the vaccines against two parasite species: Plasmodium falciparum and Toxoplasma gondii. The analysis found 18 out of the 140 parasite antigens involved with the malaria disease process. Moreover, a majority (15 out of 54) of P. falciparum parasite antigens are localized in the cell membrane. T. gondii antigens, in contrast, have a majority (19/24) of their proteins related to signaling pathways. The antigen-enriched patterns align with the life cycle stage patterns identified in our ontological parasite vaccine modeling. CONCLUSIONS The updated VO modeling and GSEA analysis capture the influence of the complex parasite life cycles and their associated antigens on vaccine development.
Collapse
Affiliation(s)
- Anthony Huffman
- Department of Computational Medicine and Bioinformatics, University of Michigan Medicine School, 48109, Ann Arbor, MI, USA
| | - Xumeng Zhang
- College of Literature, Science, and the Arts, University of Michigan, 48109, Ann Arbor, MI, USA
| | - Meghana Lanka
- College of Literature, Science, and the Arts, University of Michigan, 48109, Ann Arbor, MI, USA
| | - Jie Zheng
- Unit for Laboratory Animal Medicine, University of Michigan Medicine School, 48109, Ann Arbor, MI, USA
| | - Anna Maria Masci
- Department of Data Impact and Governance, MD Anderson Cancer Center University of Texas, TX, 77030, Houston, USA
| | - Yongqun He
- Department of Computational Medicine and Bioinformatics, University of Michigan Medicine School, 48109, Ann Arbor, MI, USA.
- Unit for Laboratory Animal Medicine, University of Michigan Medicine School, 48109, Ann Arbor, MI, USA.
- Department of Microbiology and Immunology, University of Michigan Medicine School, 48109, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Uribe-Querol E, Rosales C. Neutrophils versus Protozoan Parasites: Plasmodium, Trichomonas, Leishmania, Trypanosoma, and Entameoba. Microorganisms 2024; 12:827. [PMID: 38674770 PMCID: PMC11051968 DOI: 10.3390/microorganisms12040827] [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: 03/10/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Neutrophils are the most abundant polymorphonuclear granular leukocytes in human blood and are an essential part of the innate immune system. Neutrophils are efficient cells that eliminate pathogenic bacteria and fungi, but their role in dealing with protozoan parasitic infections remains controversial. At sites of protozoan parasite infections, a large number of infiltrating neutrophils is observed, suggesting that neutrophils are important cells for controlling the infection. Yet, in most cases, there is also a strong inflammatory response that can provoke tissue damage. Diseases like malaria, trichomoniasis, leishmaniasis, Chagas disease, and amoebiasis affect millions of people globally. In this review, we summarize these protozoan diseases and describe the novel view on how neutrophils are involved in protection from these parasites. Also, we present recent evidence that neutrophils play a double role in these infections participating both in control of the parasite and in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- Laboratorio de Biología del Desarrollo, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
11
|
Durães-Oliveira J, Palma-Marques J, Moreno C, Rodrigues A, Monteiro M, Alexandre-Pires G, da Fonseca IP, Santos-Gomes G. Chagas Disease: A Silent Threat for Dogs and Humans. Int J Mol Sci 2024; 25:3840. [PMID: 38612650 PMCID: PMC11011309 DOI: 10.3390/ijms25073840] [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: 02/06/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Chagas disease (CD) is a vector-borne Neglected Zoonotic Disease (NZD) caused by a flagellate protozoan, Trypanosoma cruzi, that affects various mammalian species across America, including humans and domestic animals. However, due to an increase in population movements and new routes of transmission, T. cruzi infection is presently considered a worldwide health concern, no longer restricted to endemic countries. Dogs play a major role in the domestic cycle by acting very efficiently as reservoirs and allowing the perpetuation of parasite transmission in endemic areas. Despite the significant progress made in recent years, still there is no vaccine against human and animal disease, there are few drugs available for the treatment of human CD, and there is no standard protocol for the treatment of canine CD. In this review, we highlight human and canine Chagas Disease in its different dimensions and interconnections. Dogs, which are considered to be the most important peridomestic reservoir and sentinel for the transmission of T. cruzi infection in a community, develop CD that is clinically similar to human CD. Therefore, an integrative approach, based on the One Health concept, bringing together the advances in genomics, immunology, and epidemiology can lead to the effective development of vaccines, new treatments, and innovative control strategies to tackle CD.
Collapse
Affiliation(s)
- João Durães-Oliveira
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Joana Palma-Marques
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Cláudia Moreno
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Armanda Rodrigues
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| | - Marta Monteiro
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Graça Alexandre-Pires
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- Centre for Interdisciplinary Research in Animal Health, CIISA, Faculty of Veterinary Medicine, FMV, University of Lisbon, ULisboa, 1649-004 Lisbon, Portugal; (G.A.-P.); (I.P.d.F.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisbon, Portugal; (J.D.-O.); (G.S.-G.)
| |
Collapse
|
12
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [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: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
13
|
Morilla MJ, Ghosal K, Romero EL. Nanomedicines against Chagas disease: a critical review. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:333-349. [PMID: 38590427 PMCID: PMC11000002 DOI: 10.3762/bjnano.15.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/12/2024] [Indexed: 04/10/2024]
Abstract
Chagas disease (CD) is the most important endemic parasitosis in South America and represents a great socioeconomic burden for the chronically ill and their families. The only currently available treatment against CD is based on the oral administration of benznidazole, an agent, developed in 1971, of controversial effectiveness on chronically ill patients and toxic to adults. So far, conventional pharmacological approaches have failed to offer more effective and less toxic alternatives to benznidazole. Nanomedicines reduce toxicity and increase the effectiveness of current oncological therapies. Could nanomedicines improve the treatment of the neglected CD? This question will be addressed in this review, first by critically discussing selected reports on the performance of benznidazole and other molecules formulated as nanomedicines in in vitro and in vivo CD models. Taking into consideration the developmental barriers for nanomedicines and the degree of current technical preclinical efforts, a prospect of developing nanomedicines against CD will be provided. Not surprisingly, we conclude that structurally simpler formulations with minimal production cost, such as oral nanocrystals and/or parenteral nano-immunostimulants, have the highest chances of making it to the market to treat CD. Nonetheless, substantive political and economic decisions, key to facing technological challenges, are still required regarding a realistic use of nanomedicines effective against CD.
Collapse
Affiliation(s)
- Maria Jose Morilla
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Kajal Ghosal
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd., Jadavpur, Kolkata 700032, West Bengal, India
| | - Eder Lilia Romero
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| |
Collapse
|
14
|
Berhe H, Kumar Cinthakunta Sridhar M, Zerihun M, Qvit N. The Potential Use of Peptides in the Fight against Chagas Disease and Leishmaniasis. Pharmaceutics 2024; 16:227. [PMID: 38399281 PMCID: PMC10892537 DOI: 10.3390/pharmaceutics16020227] [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: 11/12/2023] [Revised: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Chagas disease and leishmaniasis are both neglected tropical diseases that affect millions of people around the world. Leishmaniasis is currently the second most widespread vector-borne parasitic disease after malaria. The World Health Organization records approximately 0.7-1 million newly diagnosed leishmaniasis cases each year, resulting in approximately 20,000-30,000 deaths. Also, 25 million people worldwide are at risk of Chagas disease and an estimated 6 million people are infected with Trypanosoma cruzi. Pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine are currently used to treat leishmaniasis. Also, nifurtimox and benznidazole are two drugs currently used to treat Chagas disease. These drugs are associated with toxicity problems such as nephrotoxicity and cardiotoxicity, in addition to resistance problems. As a result, the discovery of novel therapeutic agents has emerged as a top priority and a promising alternative. Overall, there is a need for new and effective treatments for Chagas disease and leishmaniasis, as the current drugs have significant limitations. Peptide-based drugs are attractive due to their high selectiveness, effectiveness, low toxicity, and ease of production. This paper reviews the potential use of peptides in the treatment of Chagas disease and leishmaniasis. Several studies have demonstrated that peptides are effective against Chagas disease and leishmaniasis, suggesting their use in drug therapy for these diseases. Overall, peptides have the potential to be effective therapeutic agents against Chagas disease and leishmaniasis, but more research is needed to fully investigate their potential.
Collapse
Affiliation(s)
| | | | | | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel; (H.B.); (M.K.C.S.); (M.Z.)
| |
Collapse
|
15
|
Izquierdo-Condoy JS, Vásconez-Gonzáles J, Morales-Lapo E, Tello-De-la-Torre A, Naranjo-Lara P, Fernández R, Hidalgo MR, Escobar A, Yépez VH, Díaz AM, Oliva C, Ortiz-Prado E. Beyond the acute phase: a comprehensive literature review of long-term sequelae resulting from infectious diseases. Front Cell Infect Microbiol 2024; 14:1293782. [PMID: 38357446 PMCID: PMC10864624 DOI: 10.3389/fcimb.2024.1293782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
Infectious diseases have consistently served as pivotal influences on numerous civilizations, inducing morbidity, mortality, and consequently redirecting the course of history. Their impact extends far beyond the acute phase, characterized by the majority of symptom presentations, to a multitude of adverse events and sequelae that follow viral, parasitic, fungal, or bacterial infections. In this context, myriad sequelae related to various infectious diseases have been identified, spanning short to long-term durations. Although these sequelae are known to affect thousands of individuals individually, a comprehensive evaluation of all potential long-term effects of infectious diseases has yet to be undertaken. We present a comprehensive literature review delineating the primary sequelae attributable to major infectious diseases, categorized by systems, symptoms, and duration. This compilation serves as a crucial resource, illuminating the long-term ramifications of infectious diseases for healthcare professionals worldwide. Moreover, this review highlights the substantial burden that these sequelae impose on global health and economies, a facet often overshadowed by the predominant focus on the acute phase. Patients are frequently discharged following the resolution of the acute phase, with minimal long-term follow-up to comprehend and address potential sequelae. This emphasizes the pressing need for sustained vigilance, thorough patient monitoring, strategic health management, and rigorous research to understand and mitigate the lasting economic and health impacts of infectious diseases more fully.
Collapse
|
16
|
Macedo-da-Silva J, Mule SN, Rosa-Fernandes L, Palmisano G. A computational pipeline elucidating functions of conserved hypothetical Trypanosoma cruzi proteins based on public proteomic data. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 138:401-428. [PMID: 38220431 DOI: 10.1016/bs.apcsb.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The proteome is complex, dynamic, and functionally diverse. Functional proteomics aims to characterize the functions of proteins in biological systems. However, there is a delay in annotating the function of proteins, even in model organisms. This gap is even greater in other organisms, including Trypanosoma cruzi, the causative agent of the parasitic, systemic, and sometimes fatal disease called Chagas disease. About 99.8% of Trypanosoma cruzi proteome is not manually annotated (unreviewed), among which>25% are conserved hypothetical proteins (CHPs), calling attention to the knowledge gap on the protein content of this organism. CHPs are conserved proteins among different species of various evolutionary lineages; however, they lack functional validation. This study describes a bioinformatics pipeline applied to public proteomic data to infer possible biological functions of conserved hypothetical Trypanosoma cruzi proteins. Here, the adopted strategy consisted of collecting differentially expressed proteins between the epimastigote and metacyclic trypomastigotes stages of Trypanosoma cruzi; followed by the functional characterization of these CHPs applying a manifold learning technique for dimension reduction and 3D structure homology analysis (Spalog). We found a panel of 25 and 26 upregulated proteins in the epimastigote and metacyclic trypomastigote stages, respectively; among these, 18 CHPs (8 in the epimastigote stage and 10 in the metacyclic stage) were characterized. The data generated corroborate the literature and complement the functional analyses of differentially regulated proteins at each stage, as they attribute potential functions to CHPs, which are frequently identified in Trypanosoma cruzi proteomics studies. However, it is important to point out that experimental validation is required to deepen our understanding of the CHPs.
Collapse
Affiliation(s)
- Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil
| | - Simon Ngao Mule
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, NSW, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Zingales B, Macedo AM. Fifteen Years after the Definition of Trypanosoma cruzi DTUs: What Have We Learned? Life (Basel) 2023; 13:2339. [PMID: 38137940 PMCID: PMC10744745 DOI: 10.3390/life13122339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Trypanosoma cruzi, the protozoan causative of Chagas disease (ChD), exhibits striking genetic and phenotypic intraspecific diversity, along with ecoepidemiological complexity. Human-pathogen interactions lead to distinct clinical presentations of ChD. In 2009, an international consensus classified T. cruzi strains into six discrete typing units (DTUs), TcI to TcVI, later including TcBat, and proposed reproducible genotyping schemes for DTU identification. This article aims to review the impact of classifying T. cruzi strains into DTUs on our understanding of biological, ecoepidemiological, and pathogenic aspects of T. cruzi. We will explore the likely origin of DTUs and the intrinsic characteristics of each group of strains concerning genome organization, genomics, and susceptibility to drugs used in ChD treatment. We will also provide an overview of the association of DTUs with mammalian reservoirs, and summarize the geographic distribution, and the clinical implications, of prevalent specific DTUs in ChD patients. Throughout this review, we will emphasize the crucial roles of both parasite and human genetics in defining ChD pathogenesis and chemotherapy outcome.
Collapse
Affiliation(s)
- Bianca Zingales
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, São Paulo, Brazil
| | - Andréa M. Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil;
| |
Collapse
|
18
|
da Cruz Filho IJ, Duarte DMFA, Marques DSC, da Rocha JVR, Diniz EGM, Brayner FA, Alves LC, de Azevedo Albuquerque MCP, de Lima Aires A, Nogueira F, de Lima MDCA. Evaluation of the hydroalcoholic extract of Clarisia racemosa as an antiparasitic agent: an in vitro approach. 3 Biotech 2023; 13:391. [PMID: 37953832 PMCID: PMC10635994 DOI: 10.1007/s13205-023-03799-2] [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: 08/01/2023] [Accepted: 09/30/2023] [Indexed: 11/14/2023] Open
Abstract
Clarisia racemosa Ruiz & Pav is a neotropical species found in humid forests from southern Mexico to southern Brazil. There are few studies related to the ethnopharmacological use of C. racemosa. Our objective was to evaluate the hydroalcoholic extract of C. racemosa as a potential antiparasitic agent. For this, we performed in vitro assays against strains of Leishmania amazonensis, Trypanosoma cruzi, Plasmodium falciparum, and Schistosoma mansoni. At the same time, immunomodulatory activity tests were carried out. The results demonstrated that the extract was able to stimulate and activate immune cells. In preliminary antiparasitic tests, structural modifications were observed in the promastigote form of L. amazonensis and in adult worms of S. mansoni. The extract was able to inhibit the growth of trypomastigote form of T. cruzi and finally showed low antiparasitic activity against strains of P. falciparum. It is pioneering work and these results demonstrate that C. racemosa extract is a promising alternative and contributes to the arsenal of possible forms of treatment to combat parasites. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03799-2.
Collapse
Affiliation(s)
- Iranildo José da Cruz Filho
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco, Recife, PE 50670-420 Brazil
| | | | - Diego Santa Clara Marques
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco, Recife, PE 50670-420 Brazil
| | | | | | - Fábio André Brayner
- Laboratory of Immunopathology and Infectious-Parasitic Diseases Keizo Asami – LIKA, Recife, PE 50670-420 Brazil
| | - Luiz Carlos Alves
- Laboratory of Immunopathology and Infectious-Parasitic Diseases Keizo Asami – LIKA, Recife, PE 50670-420 Brazil
| | | | - André de Lima Aires
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation (IAM-FIOCRUZ), Recife, PE 50670-420 Brazil
| | - Fátima Nogueira
- Global Health and Tropical Medicine, GHTM, Institute of Hygiene and Tropical Medicine, IHMT, New University of Lisbon, 1349-008 Lisbon, Portugal
| | | |
Collapse
|
19
|
Faria AFM, de Souza Ferreira Pereira C, Teixeira GP, Dos Santos Galvão RM, Pacheco PAF, Bello ML, de Jesus DH, Calabrese K, Gonzaga DTG, Boechat N, Faria RX. In vitro evaluation of 2-(1H-pyrazol-1-yl)-1,3,4-thiadiazole derivatives against replicative and infective stages of Trypanosoma cruzi. J Bioenerg Biomembr 2023; 55:409-421. [PMID: 37919636 DOI: 10.1007/s10863-023-09982-7] [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: 05/10/2023] [Accepted: 08/07/2023] [Indexed: 11/04/2023]
Abstract
Current treatment of Chagas disease (CD) is based on two substances, nifurtimox (NT) and benzonidazole (BZ), both considered unsatisfactory mainly due to their low activities and high toxicity profile. One of the main challenges faced in CD management concerns the identification of new drugs active in the acute and chronic phases and with good pharmacokinetic profiles. In this work, we studied the bioactivity of twenty 2-(1H-pyrazol-1-yl)-1,3,4-thiadiazole derivatives against Trypanosoma cruzi epimastigotes and trypomastigotes. We identified seven derivatives with promising activity against epimastigote forms with IC50 values ranging from 6 µM to 44 µM. Most of the compounds showed no significant toxicity against murine macrophages. Our initial investigation on the mechanism of action indicates that this series of compounds may exert their anti-parasitic effect, inducing cell membrane damage. The results in trypomastigotes showed that one derivative, PDAN 78, satisfactorily inhibited metabolic alteration at all concentrations. Moreover, we used molecular modeling to understand how tridimensional and structural aspects might influence the observed bioactivities. Finally, we also used in silico approaches to assess the potential pharmacokinetic and toxicological properties of the most active compounds. Our initial results indicate that this molecular scaffold might be a valuable prototype for novel and safe trypanocidal compounds.
Collapse
Affiliation(s)
- Ana Flávia Martins Faria
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Caroline de Souza Ferreira Pereira
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Guilherme Pegas Teixeira
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Raíssa Maria Dos Santos Galvão
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Paulo Anastácio F Pacheco
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Murilo Lamim Bello
- Department of Pharmaceuticals and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daiane Hardoim de Jesus
- Laboratory of Immunomodulation and Protozoology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Kátia Calabrese
- Laboratory of Immunomodulation and Protozoology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Daniel Tadeu Gomes Gonzaga
- Department of Pharmacy, West Zone Campus, State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Nubia Boechat
- Fiocruz Institute of Drug Technology, Farmanguinhos, Fiocruz, Brazil
| | - Robson Xavier Faria
- Laboratory for Evaluation and Promotion of Evaluation and Promotion of Environmental Health (L, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Thomas M, McGonagle K, Rowland P, Robinson DA, Dodd PG, Camino-Díaz I, Campbell L, Cantizani J, Castañeda P, Conn D, Craggs PD, Edwards D, Ferguson L, Fosberry A, Frame L, Goswami P, Hu X, Korczynska J, MacLean L, Martin J, Mutter N, Osuna-Cabello M, Paterson C, Peña I, Pinto EG, Pont C, Riley J, Shishikura Y, Simeons FRC, Stojanovski L, Thomas J, Wrobel K, Young RJ, Zmuda F, Zuccotto F, Read KD, Gilbert IH, Marco M, Miles TJ, Manzano P, De Rycker M. Structure-Guided Design and Synthesis of a Pyridazinone Series of Trypanosoma cruzi Proteasome Inhibitors. J Med Chem 2023; 66:10413-10431. [PMID: 37506194 PMCID: PMC10424187 DOI: 10.1021/acs.jmedchem.3c00582] [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: 03/31/2023] [Indexed: 07/30/2023]
Abstract
There is an urgent need for new treatments for Chagas disease, a parasitic infection which mostly impacts South and Central America. We previously reported on the discovery of GSK3494245/DDD01305143, a preclinical candidate for visceral leishmaniasis which acted through inhibition of the Leishmania proteasome. A related analogue, active against Trypanosoma cruzi, showed suboptimal efficacy in an animal model of Chagas disease, so alternative proteasome inhibitors were investigated. Screening a library of phenotypically active analogues against the T. cruzi proteasome identified an active, selective pyridazinone, the development of which is described herein. We obtained a cryo-EM co-structure of proteasome and a key inhibitor and used this to drive optimization of the compounds. Alongside this, optimization of the absorption, distribution, metabolism, and excretion (ADME) properties afforded a suitable compound for mouse efficacy studies. The outcome of these studies is discussed, alongside future plans to further understand the series and its potential to deliver a new treatment for Chagas disease.
Collapse
Affiliation(s)
- Michael
G. Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kate McGonagle
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Paul Rowland
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - David A. Robinson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Peter G. Dodd
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Isabel Camino-Díaz
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Lorna Campbell
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Juan Cantizani
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pablo Castañeda
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Daniel Conn
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Peter D. Craggs
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Darren Edwards
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Liam Ferguson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Andrew Fosberry
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Laura Frame
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Panchali Goswami
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Xiao Hu
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Justyna Korczynska
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Lorna MacLean
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Julio Martin
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Nicole Mutter
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Osuna-Cabello
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Christy Paterson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Imanol Peña
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Erika G. Pinto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Caterina Pont
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Jennifer Riley
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Yoko Shishikura
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Frederick R. C. Simeons
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Laste Stojanovski
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - John Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Karolina Wrobel
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | | | - Filip Zmuda
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Fabio Zuccotto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kevin D. Read
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Ian H. Gilbert
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Marco
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Timothy J. Miles
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pilar Manzano
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Manu De Rycker
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| |
Collapse
|
21
|
Nunes Lemes LF, Magoulas GE, Souza de Oliveira A, Barrias E, de Camargo Nascente L, Granado R, Teixeira de Macedo Silva S, Assimomytis N, de Souza W, Bolognesi ML, Romeiro LAS, Calogeropoulou T. Valorizing Constituents of Cashew Nut Shell Liquid toward the Sustainable Development of New Drugs against Chagas Disease. ACS Infect Dis 2023; 9:1334-1345. [PMID: 37307287 DOI: 10.1021/acsinfecdis.3c00076] [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] [Indexed: 06/14/2023]
Abstract
Six new ether phospholipid analogues encompassing constituents from cashew nut shell liquid as the lipid portion were synthesized in an effort to valorize byproducts of the cashew industry toward the generation of potent compounds against Chagas disease. Anacardic acids, cardanols, and cardols were used as the lipid portions and choline as the polar headgroup. The compounds were evaluated for their in vitro antiparasitic activity against different developmental stages of Trypanosoma cruzi. Compounds 16 and 17 were found to be the most potent against T. cruzi epimastigotes, trypomastigotes, and intracellular amastigotes exhibiting selectivity indices against the latter 32-fold and 7-fold higher than current drug benznidazole, respectively. Hence, four out of six analogues can be considered as hit-compounds toward the sustainable development of new treatments for Chagas disease, based on inexpensive agro-waste material.
Collapse
Affiliation(s)
- Laís Flávia Nunes Lemes
- Tropical Medicine Center, Faculty of Medicine, University of Brasília, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, Distrito Federal, Brazil
- Catholic University of Brasilia, QS 07, Lote 01, EPCT, Águas Claras, 71966-700 Brasília, Distrito Federal, Brazil
| | - George E Magoulas
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Andressa Souza de Oliveira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, Distrito Federal, Brazil
| | - Emile Barrias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho s/n, Ilha do Fundão, 21941-900 Rio de Janeiro, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho s/n, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Luciana de Camargo Nascente
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, Distrito Federal, Brazil
| | - Renato Granado
- Laboratory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology - Inmetro, Rua Santa Alexandrina, 416, Rio Comprido, 20261-232 Rio de Janeiro, Brazil
| | - Sara Teixeira de Macedo Silva
- Centro Nacional de Biologia Estrutural e Bioimagem, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho s/n, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Nikos Assimomytis
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Wanderley de Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho s/n, Ilha do Fundão, 21941-900 Rio de Janeiro, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho s/n, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Luiz Antonio Soares Romeiro
- Tropical Medicine Center, Faculty of Medicine, University of Brasília, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, Distrito Federal, Brazil
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, Distrito Federal, Brazil
| | - Theodora Calogeropoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| |
Collapse
|
22
|
Marin-Neto JA, Rassi A, Oliveira GMM, Correia LCL, Ramos Júnior AN, Luquetti AO, Hasslocher-Moreno AM, Sousa ASD, Paola AAVD, Sousa ACS, Ribeiro ALP, Correia Filho D, Souza DDSMD, Cunha-Neto E, Ramires FJA, Bacal F, Nunes MDCP, Martinelli Filho M, Scanavacca MI, Saraiva RM, Oliveira Júnior WAD, Lorga-Filho AM, Guimarães ADJBDA, Braga ALL, Oliveira ASD, Sarabanda AVL, Pinto AYDN, Carmo AALD, Schmidt A, Costa ARD, Ianni BM, Markman Filho B, Rochitte CE, Macêdo CT, Mady C, Chevillard C, Virgens CMBD, Castro CND, Britto CFDPDC, Pisani C, Rassi DDC, Sobral Filho DC, Almeida DRD, Bocchi EA, Mesquita ET, Mendes FDSNS, Gondim FTP, Silva GMSD, Peixoto GDL, Lima GGD, Veloso HH, Moreira HT, Lopes HB, Pinto IMF, Ferreira JMBB, Nunes JPS, Barreto-Filho JAS, Saraiva JFK, Lannes-Vieira J, Oliveira JLM, Armaganijan LV, Martins LC, Sangenis LHC, Barbosa MPT, Almeida-Santos MA, Simões MV, Yasuda MAS, Moreira MDCV, Higuchi MDL, Monteiro MRDCC, Mediano MFF, Lima MM, Oliveira MTD, Romano MMD, Araujo NNSLD, Medeiros PDTJ, Alves RV, Teixeira RA, Pedrosa RC, Aras Junior R, Torres RM, Povoa RMDS, Rassi SG, Alves SMM, Tavares SBDN, Palmeira SL, Silva Júnior TLD, Rodrigues TDR, Madrini Junior V, Brant VMDC, Dutra WO, Dias JCP. SBC Guideline on the Diagnosis and Treatment of Patients with Cardiomyopathy of Chagas Disease - 2023. Arq Bras Cardiol 2023; 120:e20230269. [PMID: 37377258 PMCID: PMC10344417 DOI: 10.36660/abc.20230269] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Affiliation(s)
- José Antonio Marin-Neto
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Anis Rassi
- Hospital do Coração Anis Rassi , Goiânia , GO - Brasil
| | | | | | | | - Alejandro Ostermayer Luquetti
- Centro de Estudos da Doença de Chagas , Hospital das Clínicas da Universidade Federal de Goiás , Goiânia , GO - Brasil
| | | | - Andréa Silvestre de Sousa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Antônio Carlos Sobral Sousa
- Universidade Federal de Sergipe , São Cristóvão , SE - Brasil
- Hospital São Lucas , Rede D`Or São Luiz , Aracaju , SE - Brasil
| | | | | | | | - Edecio Cunha-Neto
- Universidade de São Paulo , Faculdade de Medicina da Universidade, São Paulo , SP - Brasil
| | - Felix Jose Alvarez Ramires
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Fernando Bacal
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Martino Martinelli Filho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Maurício Ibrahim Scanavacca
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Magalhães Saraiva
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Adalberto Menezes Lorga-Filho
- Instituto de Moléstias Cardiovasculares , São José do Rio Preto , SP - Brasil
- Hospital de Base de Rio Preto , São José do Rio Preto , SP - Brasil
| | | | | | - Adriana Sarmento de Oliveira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Ana Yecê das Neves Pinto
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Andre Schmidt
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Andréa Rodrigues da Costa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Barbara Maria Ianni
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Carlos Eduardo Rochitte
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Hcor , Associação Beneficente Síria , São Paulo , SP - Brasil
| | | | - Charles Mady
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Marselha - França
| | | | | | | | - Cristiano Pisani
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Evandro Tinoco Mesquita
- Hospital Universitário Antônio Pedro da Faculdade Federal Fluminense , Niterói , RJ - Brasil
| | | | | | | | | | | | - Henrique Horta Veloso
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Henrique Turin Moreira
- Hospital das Clínicas , Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , SP - Brasil
| | | | | | | | - João Paulo Silva Nunes
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Fundação Zerbini, Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | | | | | - Luiz Cláudio Martins
- Universidade Estadual de Campinas , Faculdade de Ciências Médicas , Campinas , SP - Brasil
| | | | | | | | - Marcos Vinicius Simões
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | | | | | - Maria de Lourdes Higuchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Mauro Felippe Felix Mediano
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| | - Mayara Maia Lima
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | | | | | - Renato Vieira Alves
- Instituto René Rachou , Fundação Oswaldo Cruz , Belo Horizonte , MG - Brasil
| | - Ricardo Alkmim Teixeira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Coury Pedrosa
- Hospital Universitário Clementino Fraga Filho , Instituto do Coração Edson Saad - Universidade Federal do Rio de Janeiro , RJ - Brasil
| | | | | | | | | | - Silvia Marinho Martins Alves
- Ambulatório de Doença de Chagas e Insuficiência Cardíaca do Pronto Socorro Cardiológico Universitário da Universidade de Pernambuco (PROCAPE/UPE), Recife , PE - Brasil
| | | | - Swamy Lima Palmeira
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | - Vagner Madrini Junior
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | - João Carlos Pinto Dias
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| |
Collapse
|
23
|
Osorio-Méndez JF, Téllez GA, Zapata-López D, Echeverry S, Castaño JC. Sequence analysis of SWEET transporters from trypanosomatids and evaluation of its expression in Trypanosoma cruzi. Exp Parasitol 2023; 248:108496. [PMID: 36878387 DOI: 10.1016/j.exppara.2023.108496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/31/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Trypanosoma cruzi is an obligate parasite that uses glucose as one of the main resources to maintain its survival and proliferation. In eukaryotic cells glucose transport across membranes is mediated by facilitated transport through a variety of transporters. Herein, genes from the recently described SWEET family of carbohydrate transporters were identified in trypanosomatid parasites, including the medically important species T. cruzi and Leishmania spp. The identified genes have sequences with the typical attributes of known SWEET transporters. The expression of TcSWEET, the gene for the SWEET transporter found in the T. cruzi genome, was evidenced by immunohistochemistry using a polyclonal serum raised against peptides selected from the deduced TcSWEET protein sequence. In Western blot analysis, this α-TcSWEET serum detected proteins within the theoretical molecular mass for TcSWEET (25.8 kDa) in total epimastigote lysates, suggesting its expression at this parasite stage. Additionally, this serum stained epimastigotes at localizations consistent with the cell body and the flagellum. Together, these data suggests that SWEET transporters may contribute to glucose transport in trypanosomatid parasites.
Collapse
Affiliation(s)
- Juan Felipe Osorio-Méndez
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia.
| | - Germán Alberto Téllez
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Daniela Zapata-López
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Sebastián Echeverry
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Jhon Carlos Castaño
- Grupo de Inmunología Molecular (Gymol), Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Quindío, Colombia
| |
Collapse
|
24
|
de Souza CC, de Azevedo-França JA, Barrias E, Cavalcante SCF, Vieira EG, Ferreira AMDC, de Souza W, Navarro M. Silver and copper-benznidazole derivatives as potential antiparasitic metallodrugs: Synthesis, characterization, and biological evaluation. J Inorg Biochem 2023; 239:112047. [PMID: 36428157 DOI: 10.1016/j.jinorgbio.2022.112047] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Abstract
Currently the only drug available to treat Chagas disease in Brazil is benznidazole (BZN). Therefore, there is an urgent need to discover and develop new anti- Trypanosoma cruzi candidates. In our continuous effort to enhance clinical antiparasitic drugs using synergistic strategy, BZN was coordinated to silver and copper ions to enhance its effectiveness to treat that illness. In this work, the syntheses of four novel metal-BZN complexes, [Ag(BZN)2]NO3·H2O (1), [CuCl2(BZN)(H2O)]·1/2CH3CN (2), [Ag(PPh3)2(BZN)2]NO3·H2O (3), and [Cu(PPh3)2(BNZ)2]NO3·2H2O (4), and their characterization using multiple analytical and spectroscopic techniques such as Infrared (FTIR), Nuclear Magnetic Resonance (1H, 13C, 31P), UV-Visible (UV-Vis), Electron Paramagnetic Resonance (EPR), conductivity and elemental analysis are described. IC50 (Half-maximal inhibitory concentration) values of Ag-BZN compounds are about five to ten times lower than benznidazole itself in both proliferation stages of the parasite (epimastigotes and amastigotes). The cytotoxicity of both compounds in human cells (fibroblasts and hepatocytes) are comparable to BZN, indicating that Ag-BZN complexes can be more selective than BZN.
Collapse
Affiliation(s)
- Cassiano Cunha de Souza
- Laboratório de Química Bioinorgânica e Catálise, Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
| | - José Aleixo de Azevedo-França
- Laboratório de Química Bioinorgânica e Catálise, Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Emile Barrias
- Instituto Nacional de Metrologia, Qualidade e Tecnologia, INMETRO, Xerém, RJ, Brazil
| | | | - Eduardo Guimarães Vieira
- Laboratório de Bioinorgânica, Catálise e Farmacologia, Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana Maria Da Costa Ferreira
- Laboratório de Bioinorgânica, Catálise e Farmacologia, Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Instituto Nacional de Ciência e Tecnologia (INBEB) and Centro Nacional de Biologia Estrutural e Bioimagens, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Maribel Navarro
- Laboratório de Química Bioinorgânica e Catálise, Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil.
| |
Collapse
|
25
|
Ballesteros-Casallas A, Quiroga C, Ortiz C, Benítez D, Denis PA, Figueroa D, Salas CO, Bertrand J, Tapia RA, Sánchez P, Miscione GP, Comini MA, Paulino M. Mode of action of p-quinone derivatives with trypanocidal activity studied by experimental and in silico models. Eur J Med Chem 2023; 246:114926. [PMID: 36508970 DOI: 10.1016/j.ejmech.2022.114926] [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: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Quinones are attractive pharmacological scaffolds for developing new agents for the treatment of different transmissible and non-transmissible human diseases due to their capacity to alter the cell redox homeostasis. The bioactivity and potential mode of action of 19 p-quinone derivatives fused to different aromatic rings (carbo or heterocycles) and harboring distinct substituents were investigated in infective Trypanosoma brucei brucei. All the compounds, except for a furanequinone (EC50=38 μM), proved to be similarly or even more potent (EC50 = 0.5-5.5 μM) than the clinical drug nifurtimox (EC50 = 5.3 μM). Three furanequinones and one thiazolequinone displayed a higher selectivity than nifurtimox. Two of these selective hits resulted potent inhibitors of T. cruzi proliferation (EC50=0.8-1.1 μM) but proved inactive against Leishmania infantum amastigotes. Most of the p-quinones induced a rapid and marked intracellular oxidation in T. b. brucei. DFT calculations on the oxidized quinone (Q), semiquinone (Q•-) and hydroquinone (QH2) suggest that all quinones have negative ΔG for the formation of Q•-. Qualitative and quantitative structure-activity relationship analyses in two or three dimensions of different electronic and biophysical descriptors of quinones and their corresponding bioactivities (killing potency and oxidative capacity) were performed. Charge distribution over the quinone ring carbons of Q and Q.- and the frontier orbitals energies of SUMO (Q.-) and LUMO (Q) correlate with their oxidative and trypanocidal activity. QSAR analysis also highlighted that both bromine substitution in the p-quinone ring and a bulky phenyl group attached to the furane and thiazole rings (which generates a negative charge due to the π electron system polarized by the nearby heteroatoms) are favorable for activity. By combining experimental and in silico procedures, this study disclosed important information about p-quinones that may help to rationally tune their electronic properties and biological activities.
Collapse
Affiliation(s)
- Andres Ballesteros-Casallas
- COBO, Computational Bio-Organic Chemistry, Chemistry Department, Universidad de Los Andes, Carrera 1 18A-12, Bogotá, 111711, Colombia; Bioinformatics Center, DETEMA Department, Faculty of Chemistry, Universidad de la República, General Flores 2124, Montevideo, 11600, Uruguay
| | - Cristina Quiroga
- Laboratory Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Cecilia Ortiz
- Laboratory Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Diego Benítez
- Laboratory Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Pablo A Denis
- Computational Nanotechnology, DETEMA Department, Faculty of Chemistry, Universidad de la República, General Flores 2124, Montevideo, 11600, Uruguay
| | - David Figueroa
- COBO, Computational Bio-Organic Chemistry, Chemistry Department, Universidad de Los Andes, Carrera 1 18A-12, Bogotá, 111711, Colombia
| | - Cristian O Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago, 6094411, Chile
| | - Jeanluc Bertrand
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago, 6094411, Chile
| | - Ricardo A Tapia
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago, 6094411, Chile
| | - Patricio Sánchez
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago, 6094411, Chile
| | - Gian Pietro Miscione
- COBO, Computational Bio-Organic Chemistry, Chemistry Department, Universidad de Los Andes, Carrera 1 18A-12, Bogotá, 111711, Colombia.
| | - Marcelo A Comini
- Laboratory Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay.
| | - Margot Paulino
- Bioinformatics Center, DETEMA Department, Faculty of Chemistry, Universidad de la República, General Flores 2124, Montevideo, 11600, Uruguay.
| |
Collapse
|
26
|
Identification of Aryl Polyamines Derivatives as Anti- Trypanosoma cruzi Agents Targeting Iron Superoxide Dismutase. Pharmaceutics 2022; 15:pharmaceutics15010140. [PMID: 36678771 PMCID: PMC9863987 DOI: 10.3390/pharmaceutics15010140] [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: 11/03/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
Chagas disease (CD) is a tropical and potentially fatal infection caused by Trypanosoma cruzi. Although CD was limited to Latin America as a silent disease, CD has become widespread as a result of globalization. Currently, 6-8 million people are infected worldwide, and no effective treatment is available. Here, we identify new effective agents against T. cruzi. In short, 16 aryl polyamines were screened in vitro against different T. cruzi strains, and lead compounds were evaluated in vivo after oral administration in both the acute and chronic infections. The mode of action was also evaluated at the energetic level, and its high activity profile could be ascribed to a mitochondria-dependent bioenergetic collapse and redox stress by inhibition of the Fe-SOD enzyme. We present compound 15 as a potential compound that provides a step forward for the development of new agents to combat CD.
Collapse
|
27
|
Maldonado E, Rojas DA, Urbina F, Valenzuela-Pérez L, Castillo C, Solari A. Trypanosoma cruzi DNA Polymerase β Is Phosphorylated In Vivo and In Vitro by Protein Kinase C (PKC) and Casein Kinase 2 (CK2). Cells 2022; 11:cells11223693. [PMID: 36429121 PMCID: PMC9688435 DOI: 10.3390/cells11223693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
DNA polymerase β plays a fundamental role in the life cycle of Trypanosoma cruzi since it participates in the kinetoplast DNA repair and replication. This enzyme can be found in two forms in cell extracts of T. cruzi epimastigotes form. The H form is a phosphorylated form of DNA polymerase β, while the L form is not phosphorylated. The protein kinases which are able to in vivo phosphorylate DNA polymerase β have not been identified yet. In this work, we purified the H form of this DNA polymerase and identified the phosphorylation sites. DNA polymerase β is in vivo phosphorylated at several amino acid residues including Tyr35, Thr123, Thr137 and Ser286. Thr123 is phosphorylated by casein kinase 2 and Thr137 and Ser286 are phosphorylated by protein kinase C-like enzymes. Protein kinase C encoding genes were identified in T. cruzi, and those genes were cloned, expressed in bacteria and the recombinant protein was purified. It was found that T. cruzi possesses three different protein kinase C-like enzymes named TcPKC1, TcPKC2, and TcPKC3. Both TcPKC1 and TcPKC2 were able to in vitro phosphorylate recombinant DNA polymerase β, and in addition, TcPKC1 gets auto phosphorylated. Those proteins contain several regulatory domains at the N-terminus, which are predicted to bind phosphoinositols, and TcPKC1 contains a lipocalin domain at the C-terminus that might be able to bind free fatty acids. Tyr35 is phosphorylated by an unidentified protein kinase and considering that the T. cruzi genome does not contain Tyr kinase encoding genes, it is probable that Tyr35 could be phosphorylated by a dual protein kinase. Wee1 is a eukaryotic dual protein kinase involved in cell cycle regulation. We identified a Wee1 homolog in T. cruzi and the recombinant kinase was assayed using DNA polymerase β as a substrate. T. cruzi Wee1 was able to in vitro phosphorylate recombinant DNA polymerase β, although we were not able to demonstrate specific phosphorylation on Tyr35. Those results indicate that there exists a cell signaling pathway involving PKC-like kinases in T. cruzi.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
- Correspondence: (E.M.); (A.S.)
| | - Diego A. Rojas
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Fabiola Urbina
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
| | - Lucía Valenzuela-Pérez
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
| | - Christian Castillo
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
- Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago 7500975, Chile
| | - Aldo Solari
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile
- Correspondence: (E.M.); (A.S.)
| |
Collapse
|
28
|
Sandes JM, de Figueiredo RCBQ. The endoplasmic reticulum of trypanosomatids: An unrevealed road for chemotherapy. Front Cell Infect Microbiol 2022; 12:1057774. [PMID: 36439218 PMCID: PMC9684732 DOI: 10.3389/fcimb.2022.1057774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 01/04/2024] Open
Abstract
The endoplasmic reticulum (ER) of higher eukaryotic cells forms an intricate membranous network that serves as the main processing facility for folding and assembling of secreted and membrane proteins. The ER is a highly dynamic organelle that interacts with other intracellular structures, as well as endosymbiotic pathogenic and non-pathogenic microorganisms. A strict ER quality control (ERQC) must work to ensure that proteins entering the ER are folded and processed correctly. Unfolded or misfolded proteins are usually identified, selected, and addressed to Endoplasmic Reticulum-Associated Degradation (ERAD) complex. Conversely, when there is a large demand for secreted proteins or ER imbalance, the accumulation of unfolded or misfolded proteins activates the Unfold Protein Response (UPR) to restore the ER homeostasis or, in the case of persistent ER stress, induces the cell death. Pathogenic trypanosomatids, such as Trypanosoma cruzi, Trypanosoma brucei and Leishmania spp are the etiological agents of important neglected diseases. These protozoans have a complex life cycle alternating between vertebrate and invertebrate hosts. The ER of trypanosomatids, like those found in higher eukaryotes, is also specialized for secretion, and depends on the ERAD and non-canonical UPR to deal with the ER stress. Here, we reviewed the basic aspects of ER biology, organization, and quality control in trypanosomatids. We also focused on the unusual way by which T. cruzi, T. brucei, and Leishmania spp. respond to ER stress, emphasizing how these parasites' ER-unrevealed roads might be an attractive target for chemotherapy.
Collapse
Affiliation(s)
- Jana Messias Sandes
- Laboratório de Biologia Celular e Molecular de Patógenos, Departamento de Microbiologia, Instituto Aggeu Magalhães, Recife, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Keizo Assami, Universidade Federal de Pernambuco, Recife, Brazil
| | | |
Collapse
|
29
|
Ang CW, Lee BM, Jackson CJ, Wang Y, Franzblau SG, Francisco AF, Kelly JM, Bernhardt PV, Tan L, West NP, Sykes ML, Hinton AO, Bolisetti R, Avery VM, Cooper MA, Blaskovich MA. Nitroimidazopyrazinones with Oral Activity against Tuberculosis and Chagas Disease in Mouse Models of Infection. J Med Chem 2022; 65:13125-13142. [DOI: 10.1021/acs.jmedchem.2c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chee Wei Ang
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Science, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
| | - Brendon M. Lee
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, United States
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Paul V. Bernhardt
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Melissa L. Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Alexandra O. Hinton
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Raghu Bolisetti
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A. Cooper
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark A.T. Blaskovich
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
30
|
Scariot DB, Staneviciute A, Zhu J, Li X, Scott EA, Engman DM. Leishmaniasis and Chagas disease: Is there hope in nanotechnology to fight neglected tropical diseases? Front Cell Infect Microbiol 2022; 12:1000972. [PMID: 36189341 PMCID: PMC9523166 DOI: 10.3389/fcimb.2022.1000972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Nanotechnology is revolutionizing many sectors of science, from food preservation to healthcare to energy applications. Since 1995, when the first nanomedicines started being commercialized, drug developers have relied on nanotechnology to improve the pharmacokinetic properties of bioactive molecules. The development of advanced nanomaterials has greatly enhanced drug discovery through improved pharmacotherapeutic effects and reduction of toxicity and side effects. Therefore, highly toxic treatments such as cancer chemotherapy, have benefited from nanotechnology. Considering the toxicity of the few therapeutic options to treat neglected tropical diseases, such as leishmaniasis and Chagas disease, nanotechnology has also been explored as a potential innovation to treat these diseases. However, despite the significant research progress over the years, the benefits of nanotechnology for both diseases are still limited to preliminary animal studies, raising the question about the clinical utility of nanomedicines in this field. From this perspective, this review aims to discuss recent nanotechnological developments, the advantages of nanoformulations over current leishmanicidal and trypanocidal drugs, limitations of nano-based drugs, and research gaps that still must be filled to make these novel drug delivery systems a reality for leishmaniasis and Chagas disease treatment.
Collapse
Affiliation(s)
- Debora B. Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
- *Correspondence: Debora B. Scariot,
| | - Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Jennifer Zhu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Northwestern University, Chicago, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - David M. Engman
- Department of Pathology, Northwestern University, Chicago, IL, United States
| |
Collapse
|