1
|
Lye LF, Dobson DE, Beverley SM, Tung MC. RNA interference in protozoan parasites and its application. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:281-287. [PMID: 39884870 DOI: 10.1016/j.jmii.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/24/2024] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
RNA interference (RNAi) is a biological process in which RNA molecules are involved in sequence-specific suppression of gene expression, via small RNA triggers derived from double-stranded RNA that can target specific genes; it is a natural process that plays a role in both the regulation of protein synthesis and in immunity. Discovery of RNAi by Fire and Mello in 1998 had a profound impact on unraveling novel aspects of eukaryotic biology. RNA interference (RNAi) has proven to be an immensely useful tool for studying gene function and validation of potential drug targets in almost all organisms. A great advance in parasitic protozoa was achieved by the experimental demonstration of RNAi in Trypanosoma brucei, and in other protists such as Leishmania braziliensis, Entamoeba histolytica and Giardia lamblia/intestinalis. These organisms exhibit numerous differences beyond the core 'dicer' and 'slicer' activities, thereby expanding knowledge of the evolutionary diversification of this pathway in eukaryotes. When present, RNAi has led to new technologies for engineering powerful and facile knockdowns in gene expression, revolutionizing biomedical research and opening clinical potentialities. In this review, we discuss the distribution of RNAi pathways, their biological roles, and experimental applications in protozoan parasites.
Collapse
Affiliation(s)
- Lon-Fye Lye
- Department of Medical Research, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan.
| | - Deborah E Dobson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Stephen M Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Min-Che Tung
- Department of Urology, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan.
| |
Collapse
|
2
|
Bellini NK, de Lima PLC, Pires DDS, da Cunha JPC. Hidden origami in Trypanosoma cruzi nuclei highlights its non-random 3D genomic organization. mBio 2025; 16:e0386124. [PMID: 40243368 PMCID: PMC12077095 DOI: 10.1128/mbio.03861-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The protozoan Trypanosoma cruzi is the causative agent of Chagas disease and is known for its polycistronic transcription, with about 50% of its genome consisting of repetitive sequences, including coding (primarily multigenic families) and non-coding regions (such as ribosomal DNA, spliced leader [SL], and retroelements, etc). Here, we evaluated the genomic features associated with higher-order chromatin organization in T. cruzi (Brazil A4 strain) by extensive computational processing of high-throughput chromosome conformation capture (Hi-C). Through the mHi-C pipeline, designed to handle multimapping reads, we demonstrated that applying canonical Hi-C processing, which overlooks repetitive DNA sequences, results in a loss of DNA-DNA contacts, misidentifying them as chromatin-folding (CF) boundaries. Our analysis revealed that loci encoding multigenic families of virulence factors are enriched in chromatin loops and form shorter and tighter CF domains than the loci encoding core genes. We uncovered a non-random three-dimensional (3D) genomic organization in which nonprotein-coding RNA loci (transfer RNAs [tRNAs], small nuclear RNAs, and small nucleolar RNAs) and transcription termination sites are preferentially located at the boundaries of the CF domains. Our data indicate 3D clustering of tRNA loci, likely optimizing transcription by RNA polymerase III, and a complex interaction between spliced leader RNA and 18S rRNA loci, suggesting a link between RNA polymerase I and II machineries. Finally, we highlighted a group of genes encoding virulence factors that interact with SL-RNA loci, suggesting a potential regulatory role. Our findings provide insights into 3D genome organization in T. cruzi, contributing to the understanding of supranucleosomal-level chromatin organization and suggesting possible links between 3D architecture and gene expression.IMPORTANCEDespite the knowledge about the linear genome sequence and the identification of numerous virulence factors in the protozoan parasite Trypanosoma cruzi, there has been a limited understanding of how these genomic features are spatially organized within the nucleus and how this organization impacts gene regulation and pathogenicity. By providing a detailed analysis of the three-dimensional (3D) chromatin architecture in T. cruzi, our study contributed to narrowing this gap. We deciphered part of the origami structure hidden in the T. cruzi nucleus, showing the unidimensional genomic features are non-randomly 3D organized in the nuclear organelle. We uncovered the role of nonprotein-coding RNA loci (e.g., transfer RNAs, spliced leader RNA, and 18S RNA) in shaping genomic architecture, offering insights into an additional epigenetic layer that may influence gene expression.
Collapse
Affiliation(s)
- Natália Karla Bellini
- Cell Cycle Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| | - Pedro Leonardo Carvalho de Lima
- Cell Cycle Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| | - David da Silva Pires
- Cell Cycle Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| | - Julia Pinheiro Chagas da Cunha
- Cell Cycle Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, Brazil
| |
Collapse
|
3
|
Carlier Y, Dumonteil E, Herrera C, Waleckx E, Tibayrenc M, Buekens P, Truyens C, Muraille E. Coinfection by multiple Trypanosoma cruzi clones: a new perspective on host-parasite relationship with consequences for pathogenesis and management of Chagas disease. Microbiol Mol Biol Rev 2025:e0024224. [PMID: 40116484 DOI: 10.1128/mmbr.00242-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
SUMMARYChagas disease (CD) is caused by the protozoan parasite Trypanosoma cruzi (Tc), infecting 6-7 million people. It is transmitted by insect vectors, orally, through infected tissues, or congenitally. Tc infection can progress toward chronic cardiac and/or digestive severe and fatal CD in 20%-40% of patients. Tc exhibits an important genetic and phenotypic intraspecies diversity and a preponderant clonal population structure. The impact of multiclonal coinfections has been little studied in CD patients. Relationships between the currently used discrete typing unit (DTU)-based classification of Tc lineages and the occurrence of the different clinical forms of CD, its congenital transmission, as well as the efficacy of trypanocidal molecules (benznidazole and nifurtimox) could not be established. In this review, we revisit the different aspects of Tc diversity and analyze the impact of infections with multiple clones and their variants on the dynamic and pathogenesis of CD and its maternal-fetal transmission. We propose to call "cruziome" all the Tc clones and their variants infecting a given host and provide strong evidence that (i) multiclonal Tc infections are likely the rule rather than the exception; (ii) each "cruziome" is associated with a unique combination of virulence factors, tissular tropisms, and host immune responses; (iii) accordingly, some particularly harmful "cruziomes" likely trigger the occurrence and progression of CD and might also favor the congenital transmission of parasites. We propose that our concept of "cruziome" should be taken into consideration because of its practical consequences in epidemiological studies, laboratory diagnosis, clinical management, and treatment of CD.
Collapse
Affiliation(s)
- Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Tropical Medicine and Infectious Disease, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Eric Dumonteil
- Department of Tropical Medicine and Infectious Disease, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Claudia Herrera
- Department of Tropical Medicine and Infectious Disease, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Etienne Waleckx
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán (UADY), Mérida, Mexico
- Institut de Recherche pour le Développement (IRD), UMR INTERTRYP, IRD, CIRAD, Université de Montpellier, Montpellier, France
| | - Michel Tibayrenc
- Institut de Recherche pour le Développement (IRD), UMR INTERTRYP, IRD, CIRAD, Université de Montpellier, Montpellier, France
| | - Pierre Buekens
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Carine Truyens
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Unité de Recherche en Biologie des Microorganismes (URBM), Laboratoire d'Immunologie et de Microbiologie (NARILIS), Université de Namur, Namur, Belgium
| |
Collapse
|
4
|
Spaulding A, Sharma A, Giardini MA, Hoffman B, Bernatchez JA, McCall LI, Calvet CM, Ackermann J, Souza JM, Thomas D, Millard CC, Devine WG, Singh B, Silva EM, Leed SE, Roncal NE, Penn EC, Erath J, Kumar G, Sepulveda Y, Garcia A, Rodriguez A, El-Sakkary N, Sciotti RJ, Campbell RF, Momper JD, McKerrow JH, Caffrey CR, Siqueira-Neto JL, Pollastri MP, Mensa-Wilmot K, Ferrins L. Identification of Substituted 4-Aminocinnolines as Broad-Spectrum Antiparasitic Agents. ACS Infect Dis 2025; 11:584-599. [PMID: 39936822 PMCID: PMC11915375 DOI: 10.1021/acsinfecdis.4c00666] [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: 08/22/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
Neglected tropical diseases such as Chagas disease, human African trypanosomiasis, leishmaniasis, and schistosomiasis have a significant global health impact in predominantly developing countries, although these diseases are spreading due to increased international travel and population migration. Drug repurposing with a focus on increasing antiparasitic potency and drug-like properties is a cost-effective and efficient route to the development of new therapies. Here we identify compounds that have potent activity against Trypanosoma cruzi and Leishmania donovani, and the latter were progressed into the murine model of infection. Despite the potent in vitro activity, there was no effect on parasitemia, necessitating further work to improve the pharmacokinetic properties of this series. Nonetheless, valuable insights have been obtained into the structure-activity and structure-property relationships of this compound series.
Collapse
Affiliation(s)
- Andrew Spaulding
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Amrita Sharma
- Department
of Molecular and Cellular Biology, Kennesaw
State University, Science
Building MailDrop1201, 370 Paulding Avenue, Kennesaw, Georgia 30144, United States
| | - Miriam A. Giardini
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Benjamin Hoffman
- Department
of Molecular and Cellular Biology, Kennesaw
State University, Science
Building MailDrop1201, 370 Paulding Avenue, Kennesaw, Georgia 30144, United States
| | - Jean A. Bernatchez
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Laura-Isobel McCall
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Claudia M. Calvet
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Jasmin Ackermann
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Julia M. Souza
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Diane Thomas
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Caroline C. Millard
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - William G. Devine
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Baljinder Singh
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Everton M. Silva
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Susan E. Leed
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Norma E. Roncal
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Erica C. Penn
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Jessey Erath
- Department
of Microbiology, New York University School
of Medicine, 430 E. 29th Street, New York, New York 10010, United States
| | - Gaurav Kumar
- Department
of Molecular and Cellular Biology, Kennesaw
State University, Science
Building MailDrop1201, 370 Paulding Avenue, Kennesaw, Georgia 30144, United States
| | - Yadira Sepulveda
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Arnold Garcia
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Ana Rodriguez
- Department
of Microbiology, New York University School
of Medicine, 430 E. 29th Street, New York, New York 10010, United States
| | - Nelly El-Sakkary
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Richard J. Sciotti
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Robert F. Campbell
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Jeremiah D. Momper
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - James H. McKerrow
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Conor R. Caffrey
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Jair L. Siqueira-Neto
- Center for
Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy
and Pharmaceutical Sciences, University
of California, San Diego, La Jolla, California 92093, United States
| | - Michael P. Pollastri
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Kojo Mensa-Wilmot
- Department
of Molecular and Cellular Biology, Kennesaw
State University, Science
Building MailDrop1201, 370 Paulding Avenue, Kennesaw, Georgia 30144, United States
| | - Lori Ferrins
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
- Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Pal S, Dam S. Identification and characterisation of a novel EhOrc1/Cdc6 from the human pathogen Entamoeba histolytica: an in silico approach. J Biomol Struct Dyn 2025; 43:1883-1892. [PMID: 38095553 DOI: 10.1080/07391102.2023.2293264] [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: 08/02/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2025]
Abstract
The onset of a pre-replication complex on origin commences DNA replication. The Origin recognition complex (Orc), Cell division cycle protein 6 (Cdc6), and the minichromosome maintenance (Mcm) replicative helicase, along with Chromatin licensing and DNA replication factor 1 (Cdt1), make up the pre-replication complex in eukaryotes. Eukaryotic Orc is made up of six subunits, designated Orc1-6 while monomeric Cdc6 has sequence similarity with Orc1. However, Orc has remained unexplored in the protozoan parasite Entamoeba histolytica. Here we report a single functional Orc1/Cdc6 protein in E. histolytica. Its structural and functional aspects have been highlighted by a detailed in silico analysis that reflects physicochemical characteristics, predictive 3D structure modelling, protein-protein interaction studies, molecular docking and simulation. This in silico study provides insight into EhOrc1/Cdc6 and points out that E. histolytica carries pre-replication machinery that is less complex than higher eukaryotes and closer to archaea. Additionally, it lays the groundwork for future investigations into the methods of origin recognition, and anomalies of cell cycle observed in this enigmatic parasite.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suchetana Pal
- Department of Microbiology, The University of Burdwan, Burdwan, West Bengal, India
| | - Somasri Dam
- Department of Microbiology, The University of Burdwan, Burdwan, West Bengal, India
| |
Collapse
|
6
|
Berná L, Chiribao ML, Pita S, Alvarez-Valin F, Parodi-Talice A. Exploring the genomic landscape of the GP63 family in Trypanosoma cruzi: Evolutionary dynamics and functional peculiarities. PLoS Negl Trop Dis 2025; 19:e0012950. [PMID: 40096186 PMCID: PMC11957388 DOI: 10.1371/journal.pntd.0012950] [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: 06/03/2024] [Revised: 03/31/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Members of the GP63 metalloprotease family play crucial roles in parasite-host interactions, immune evasion, and pathogenesis. Although it has been widely studied in Leishmania spp., less is known about its function and diversity in Trypanosoma cruzi. This study focuses on characterizing the complete repertoire of GP63 sequences in the T. cruzi genome, refining gene annotations, and exploring the evolutionary dynamics that shape the diversity of these proteins. Eleven GP63 groups were identified, which are sharply defined and have a higher intra- than inter-group sequence identity. These GP63 groups display some distinctive features. First, two groups lack an essential amino acid in the active site, indicating that they are enzymatically inactive. Second, GP63 groups show strong preference for different genomic compartments. Moreover, genes from groups located in the core genome compartment of T. cruzi, are often arranged as tandem arrays (of larger genomic fragments that generally include a SIRE retroposon), whereas genes from groups located in the disruptive compartment tend to be surrounded by genes encoding other surface proteins (such as MASP, mucins and trans-sialidases). Transcription patterns across different life cycle stages are not homogenous. Instead, some GP63 groups have higher mRNA levels in the infective trypomastigote stage, suggesting a potential role in host invasion. To get a wider picture of the evolutionary dynamics of these proteins, a phylogenetic analysis was conducted that included species representative of kinetoplastid diversity. It was found that 10 out of 11 GP63 T. cruzi groups are specific to the Trypanosoma genus, suggesting that the diversification of these subfamilies took place before speciation of the genus, followed by other species-specific expansions. Additionally, there are other GP63 groups that are absent in T. cruzi. Notably, the processes of expansion and diversification of GP63 in Leishmania is independent of that of trypanosomes. This suggests that these proteins may have evolved under species-specific selective (functional) pressures, resulting in unique amplifications in each parasite species.
Collapse
Affiliation(s)
- Luisa Berná
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo, Uruguay
- Laboratorio de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Laura Chiribao
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Pita
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo, Uruguay
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Fernando Alvarez-Valin
- Laboratorio de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Adriana Parodi-Talice
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo, Uruguay
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
7
|
Agyabeng-Dadzie F, Beaudry MS, Deyanov A, Slanis H, Duong MQ, Turner R, Khan A, Arias CA, Kissinger JC, Glenn TC, de Paula Baptista R. Evaluating the Benefits and Limits of Multiple Displacement Amplification With Whole-Genome Oxford Nanopore Sequencing. Mol Ecol Resour 2025:e14094. [PMID: 40018988 DOI: 10.1111/1755-0998.14094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Multiple displacement amplification (MDA) outperforms conventional PCR in long fragment and whole-genome amplification, making it attractive to couple MDA with long-read sequencing of samples with limited quantities of DNA to obtain improved genome assemblies. Here, we explore the efficacy and limits of MDA for efficient low-cost genome sequence assembly using Oxford Nanopore Technologies (ONTs) rapid library preparations and minION sequencing. We successfully generated almost complete genome sequences for all organisms examined, including Gram-positive (Staphylococcus aureus, Enterococcus faecium) and Gram-negative (Escherichia coli) prokaryotes and one challenging eukaryotic pathogen (Cryptosporidium spp) representing a broad spectrum of critical infectious disease pathogens. High-quality data from those samples were generated starting with only 0.025 ng of total DNA. Controlled sheared DNA samples exhibited a distinct pattern of size increase after MDA, which may be associated with the amplification of long, low-abundance fragments present in the assay, as well as generating concatemeric sequences during amplification. To address concatemers, we developed a computational pipeline (CADECT: Concatemer Detection Tool) to identify and remove putative concatemeric sequences. This study highlights the efficacy of MDA in generating high-quality genome assemblies from limited amounts of input DNA. Also, the CADECT pipeline effectively mitigated the impact of concatemeric sequences, enabling the assembly of contiguous sequences even in cases where the input genomic DNA was degraded. These results have significant implications for the study of organisms that are challenging to culture in vitro, such as Cryptosporidium, and for expediting critical results in clinical settings with limited quantities of available genomic DNA.
Collapse
Affiliation(s)
| | - Megan S Beaudry
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - Alex Deyanov
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, Texas, USA
| | - Haley Slanis
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, Texas, USA
| | - Minh Q Duong
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, Texas, USA
| | - Randi Turner
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- USA Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Service, Animal Parasitic Disease Laboratory, Beltsville, Maryland, USA
| | - Asis Khan
- USA Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Service, Animal Parasitic Disease Laboratory, Beltsville, Maryland, USA
| | - Cesar A Arias
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, Texas, USA
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jessica C Kissinger
- Department of Genetics, University of Georgia, Athens, Georgia, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Travis C Glenn
- Department of Genetics, University of Georgia, Athens, Georgia, USA
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Rodrigo de Paula Baptista
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, Texas, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
8
|
Wiedeman J, Harrison R, Etheridge RD. A limitation lifted: A conditional knockdown system reveals essential roles for Polo-like kinase and Aurora kinase 1 in Trypanosoma cruzi cell division. Proc Natl Acad Sci U S A 2025; 122:e2416009122. [PMID: 40106484 PMCID: PMC11874021 DOI: 10.1073/pnas.2416009122] [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: 08/07/2024] [Accepted: 12/20/2024] [Indexed: 03/22/2025] Open
Abstract
While advances in genome editing technologies have simplified gene disruption in many organisms, the study of essential genes requires development of conditional disruption or knockdown systems that are not available in most organisms. Such is the case for Trypanosoma cruzi, a parasite that causes Chagas disease, a severely neglected tropical disease endemic to Latin America that is often fatal. Our knowledge of the identity of essential genes and their functions in T. cruzi has been severely constrained by historical challenges in very basic genetic manipulation and the absence of RNA interference machinery. Here, we describe the development and use of self-cleaving RNA sequences to conditionally regulate essential gene expression in T. cruzi. Using these tools, we identified essential roles for Polo-like and Aurora kinases in T. cruzi cell division, mirroring their functions in Trypanosoma brucei. Importantly, we demonstrate conditional knockdown of essential genes in intracellular amastigotes, the disease-causing stage of the parasite in its human host. This conditional knockdown system enables the efficient and scalable functional characterization of essential genes in T. cruzi and provides a framework for the development of conditional gene knockdown systems for other nonmodel organisms.
Collapse
Affiliation(s)
- Justin Wiedeman
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30602
- Department of Cellular Biology, University of Georgia, Athens, GA30602
| | - Ruby Harrison
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30602
- Department of Cellular Biology, University of Georgia, Athens, GA30602
| | - Ronald Drew Etheridge
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30602
- Department of Cellular Biology, University of Georgia, Athens, GA30602
| |
Collapse
|
9
|
Dean AAC, Berná L, Robello C, Buscaglia CA, Balouz V. An algorithm for annotation and classification of T. cruzi MASP sequences: towards a better understanding of the parasite genetic variability. BMC Genomics 2025; 26:194. [PMID: 39994548 PMCID: PMC11852901 DOI: 10.1186/s12864-025-11384-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Trypanosoma cruzi, the protozoan causing Chagas disease, is responsible for a neglected tropical disease affecting millions in Latin America. Its genome contains rapidly evolving multigene families, such as mucins (TcMUC), trans-sialidases (TS), and mucin-associated surface proteins (MASP), which are essential for parasite transmission and disease mechanisms. However, methodological challenges in genome assembly and annotation have limited the characterization of these gene families, particularly MASPs. RESULTS We developed a bioinformatic pipeline for the automatic identification, characterization, and annotation of MASPs directly from T. cruzi genome assemblies. This algorithm, based on a manually curated MASP database and HMM-based identification of MASP diagnostic motifs, enables the robust classification of these molecules into canonical MASPs, MASP-related molecules (mostly pseudogenes), and chimeric sequences combining MASPs and TcMUC/TS genes. Validation against a rigorously annotated dataset demonstrated high accuracy, and allowed us to reclassify misanotated sequences and, more crucially, to accurately identify previously unrecognized canonical MASPs and MASP chimeras. This algorithm was then used to explore the MASP repertoire in the genomes of 13 parasite strains from different evolutionary lineages, revealing patterns of diversity. For instance, TcI and TcII strains exhibited higher ratios of canonical MASP/MASP-related molecules and a greater abundance of MASP chimeras, suggesting that their genomes are under strong selective pressures towards maintaining a broader panel of full-length MASP genes at the expense of pseudogenes. On the contrary, structural features of canonical MASPs, MASP-related sequences, and MASP-chimeras were largely conserved across parasite genomes. CONCLUSIONS This novel pipeline automates the annotation of MASPs, a key surface protein family unique to T. cruzi, improving genome annotation and enabling robust comparative analyses. It provides an essential tool for exploring the evolutionary dynamics of multigene families in T. cruzi and could be extended to other gene families.
Collapse
Affiliation(s)
- Aldana Alexandra Cepeda Dean
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. 25 de Mayo y Francia, Campus UNSAM, B1650HMP San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), UNSAM, Buenos Aires, Argentina
| | - Luisa Berná
- Laboratorio de Interacciones Hospedero-Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Laboratorio de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero-Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Unidad Académica de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Carlos Andrés Buscaglia
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. 25 de Mayo y Francia, Campus UNSAM, B1650HMP San Martín, Buenos Aires, Argentina.
- Escuela de Bio y Nanotecnologías (EByN), UNSAM, Buenos Aires, Argentina.
| | - Virginia Balouz
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. 25 de Mayo y Francia, Campus UNSAM, B1650HMP San Martín, Buenos Aires, Argentina.
- Escuela de Bio y Nanotecnologías (EByN), UNSAM, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Kortbawi HM, Marczak RJ, Rajan JV, Bulaong NL, Pak JE, Wu W, Wang G, Mitchell A, Saxena A, Maheshwari A, Fleischmann CJ, Kelly EA, Teal E, Townsend RL, Stramer SL, Okamoto EE, Sherbuk JE, Clark EH, Gilman RH, Colanzi R, Gennatas ED, Bern C, DeRisi JL, Whitman JD. A Trypanosoma cruzi Trans-Sialidase Peptide Demonstrates High Serological Prevalence Among Infected Populations Across Endemic Regions of Latin America. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.22.25320967. [PMID: 39974016 PMCID: PMC11838992 DOI: 10.1101/2025.01.22.25320967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Infection by Trypanosoma cruzi, the agent of Chagas disease, can irreparably damage the cardiac and gastrointestinal systems during decades of parasite persistence and related inflammation in these tissues. Diagnosis of chronic disease requires confirmation by multiple serological assays due to the imperfect performance of existing clinical tests. Current serology tests utilize antigens discovered over three decades ago with small specimen sets predominantly from South America, and lower test performance has been observed in patients who acquired T. cruzi infection in Central America and Mexico. Here, we attempt to address this gap by evaluating antibody responses against the entire T. cruzi proteome with phage display immunoprecipitation sequencing comprised of 228,127 47-amino acid peptides. We utilized diverse specimen sets from Mexico, Central America and South America, as well as different stages of cardiac disease severity, from 185 cases and 143 controls. We identified over 1,300 antigenic T. cruzi peptides derived from 961 proteins between specimen sets. A total of 67 peptides were reactive in 70% of samples across all regions, and 3 peptide epitopes were enriched in ≥90% of seropositive samples. Of these three, only one antigen, belonging to the trans-sialidase family, has not previously been described as a diagnostic target. Orthogonal validation of this peptide demonstrated increased antibody reactivity for infections originating from Central America. Overall, this study provides proteome-wide identification of seroreactive T. cruzi peptides across a large cohort spanning multiple endemic areas and identified a novel trans-sialidase peptide antigen (TS-2.23) with significant potential for translation into diagnostic serological assays.
Collapse
Affiliation(s)
- Hannah M. Kortbawi
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco; San Francisco, CA, USA
| | - Ryan J. Marczak
- Department of Epidemiology and Biostatistics, University of California San Francisco; San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Jayant V. Rajan
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Nash L. Bulaong
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - John E. Pak
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Wesley Wu
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Grace Wang
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Anthea Mitchell
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Aditi Saxena
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Aditi Maheshwari
- Department of Epidemiology and Biostatistics, University of California San Francisco; San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Charles J. Fleischmann
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Emily A. Kelly
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Evan Teal
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| | | | | | - Emi E. Okamoto
- New York University School of Medicine; New York, NY, USA
| | | | - Eva H. Clark
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine; Houston, TX, USA
- Division of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine; Houston, TX, USA
| | - Robert H. Gilman
- Johns Hopkins University Bloomberg School of Public Health; Baltimore, MD, USA
| | - Rony Colanzi
- Universidad Catolica Boliviana; Santa Cruz, Plurinational State of Bolivia
| | - Efstathios D. Gennatas
- Department of Epidemiology and Biostatistics, University of California San Francisco; San Francisco, CA, USA
- Department of Medicine, University of California San Francisco; San Francisco, CA, USA
| | - Caryn Bern
- Department of Epidemiology and Biostatistics, University of California San Francisco; San Francisco, CA, USA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA, USA
| | - Jeffrey D. Whitman
- Department of Laboratory Medicine, University of California, San Francisco; San Francisco, CA, USA
| |
Collapse
|
11
|
Ramírez-Delgado D, Flores-López CA. Molecular Markers for the Phylogenetic Reconstruction of Trypanosoma cruzi: A Quantitative Review. Pathogens 2025; 14:72. [PMID: 39861033 PMCID: PMC11768369 DOI: 10.3390/pathogens14010072] [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: 11/27/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Trypanosoma cruzi is the parasite responsible for Chagas disease, which has a significant amount of genetic diversification among the species complex. Many efforts are routinely made to characterize the genetic lineages of T. cruzi circulating in a particular geographic area. However, the genetic loci used to typify the genetic lineages of T. cruzi have not been consistent between studies. We report a quantitative analysis of the phylogenetic power that is acquired from the commonly used genetic loci that are employed for the typification of T. cruzi into its current taxonomic nomenclature. Based on three quantitative criteria (the number of phylogenetic informative characters, number of available reference sequences in public repositories, and accessibility to DNA sequences for their use as outgroup sequences), we examine and discuss the most appropriate genetic loci for the genetic typification of T. cruzi. Although the mini-exon gene is by far the locus that has been most widely used, it is not the most appropriate marker for the typification of T. cruzi based on the construction of a resolved phylogenetic tree. Overall, the mitochondrial COII-NDI locus stands out as the best molecular marker for this purpose, followed by the Cytochrome b and the Lathosterol oxidase genes.
Collapse
|
12
|
Herreros-Cabello A, Callejas-Hernández F, Gironès N, Fresno M. Trypanosoma cruzi: Genomic Diversity and Structure. Pathogens 2025; 14:61. [PMID: 39861022 PMCID: PMC11768934 DOI: 10.3390/pathogens14010061] [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: 12/12/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, a neglected tropical disease, and one of the most important parasitic diseases worldwide. The first genome of T. cruzi was sequenced in 2005, and its complexity made assembly and annotation challenging. Nowadays, new sequencing methods have improved some strains' genome sequence and annotation, revealing this parasite's extensive genetic diversity and complexity. In this review, we examine the genetic diversity, the genomic structure, and the principal multi-gene families involved in the pathogenicity of T. cruzi. The T. cruzi genome sequence is divided into two compartments: the core (conserved) and the disruptive (variable in length and multicopy gene families among strains). The disruptive region has also been described as genome plasticity and plays a key role in the parasite survival and infection process. This region comprises several multi-gene families, including trans-sialidases, mucins, and mucin-associated surface proteins (MASPs). Trans-sialidases are the most prevalent genes in the genome with a key role in the infection process, while mucins and MASPs are also significant glycosylated proteins expressed on the parasite surface, essential for its biological functions, as host-parasite interaction, host cell invasion or protection against the host immune system, in both insect and mammalian stages. Collectively, in this review, some of the most recent advances in the structure and composition of the T. cruzi genome are reviewed.
Collapse
Affiliation(s)
- Alfonso Herreros-Cabello
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | | | - Núria Gironès
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
- Instituto Sanitario de Investigación Princesa, 28006 Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
- Instituto Sanitario de Investigación Princesa, 28006 Madrid, Spain
| |
Collapse
|
13
|
Gabiatti B, Freire E, Ferreira da Costa J, Ferrarini M, Reichert Assunção de Matos T, Preti H, Munhoz da Rocha I, Guimarães B, Kramer S, Zanchin N, Holetz F. Trypanosoma cruzi eIF4E3- and eIF4E4-containing complexes bind different mRNAs and may sequester inactive mRNAs during nutritional stress. Nucleic Acids Res 2025; 53:gkae1181. [PMID: 39658061 PMCID: PMC11754739 DOI: 10.1093/nar/gkae1181] [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: 04/23/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Many eIF4F and poly(A)-binding protein (PABP) paralogues are found in trypanosomes: six eIF4E, five eIF4G, one eIF4A and two PABPs. They are expressed simultaneously and assemble into different complexes, contrasting the situation in metazoans that use distinct complexes in different cell types/developmental stages. Each eIF4F complex has its own proteins, messenger RNAs (mRNAs) and, consequently, a distinct function. We set out to study the function and regulation of the two eIF4F complexes of the parasite Trypanosoma cruzi and identified the associated proteins and mRNAs of eIF4E3 and eIF4E4 in cells in exponential growth and in nutritional stress, an inducer of differentiation to an infective stage. Upon stress, eIF4G and PABP remain associated with the eIF4E, but the associations with other 43S pre-initiation factors decrease, indicating ribosome attachment is impaired. Most eIF4E3-associated mRNAs encode for proteins involved in anabolic metabolism, while eIF4E4 associate with mRNAs encoding ribosomal proteins as in Trypanosoma brucei. Interestingly, for both eIF4E3/4, more mRNAs were associated in stressed cells than in non-stressed cells, even though these have lower translational efficiencies in stress. In summary, trypanosomes have two co-existing eIF4F complexes associating to different mRNAs, but not stress/differentiation-associated mRNAs. Under stress, both complexes exit translation but remain bound to their mRNA targets.
Collapse
Affiliation(s)
- Bernardo Papini Gabiatti
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
- Biocenter, University of Würzburg, Am Hubland 97074, Würzburg, Germany
| | - Eden Ribeiro Freire
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Jimena Ferreira da Costa
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Mariana Galvão Ferrarini
- Laboratoire de Biométrie et Biologie Évolutive, UMR 5558, CNRS, Université de Lyon, Université Lyon 1, Villeurbanne, France
| | | | - Henrique Preti
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Isadora Munhoz da Rocha
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Beatriz Gomes Guimarães
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Susanne Kramer
- Biocenter, University of Würzburg, Am Hubland 97074, Würzburg, Germany
| | - Nilson Ivo Tonin Zanchin
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| | - Fabíola Barbieri Holetz
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ, R. Prof. Algacyr Munhoz Mader 3775, 81350-010, Curitiba-PR, Brazil
| |
Collapse
|
14
|
Nájera CA, Soares-Silva M, Maeda FY, DaRocha WD, Meneghelli I, Mendes AC, Batista MF, Silva CV, da Silveira JF, Orikaza CM, Yoshida N, Silva VG, Teixeira SMR, Bartholomeu DC, Bahia D. Trypanosoma cruzi Vps34 colocalizes with Beclin1 and plays a role in parasite invasion of the host cell by modulating the expression of a sub-group of trans-sialidases. Microbes Infect 2024; 26:105385. [PMID: 38950642 DOI: 10.1016/j.micinf.2024.105385] [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: 12/19/2023] [Revised: 05/13/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Trypanosoma cruzi, the etiological agent of Chagas' disease, can infect both phagocytic and non-phagocytic cells. T. cruzi gp82 and gp90 are cell surface proteins belonging to Group II trans-sialidases known to be involved in host cell binding and invasion. Phosphatidylinositol kinases (PIK) are lipid kinases that phosphorylate phospholipids in their substrates or in themselves, regulating important cellular functions such as metabolism, cell cycle and survival. Vps34, a class III PIK, regulates autophagy, trimeric G-protein signaling, and the mTOR (mammalian Target of Rapamycin) nutrient-sensing pathway. The mammalian autophagy gene Beclin1 interacts to Vps34 forming Beclin 1-Vps34 complexes involved in autophagy and protein sorting. In T. cruzi epimastigotes, (a non-infective replicative form), TcVps34 has been related to morphological and functional changes associated to vesicular trafficking, osmoregulation and receptor-mediated endocytosis. We aimed to characterize the role of TcVps34 during invasion of HeLa cells by metacyclic (MT) forms. MTs overexpressing TcVps34 showed lower invasion rates compared to controls, whilst exhibiting a significant decrease in gp82 expression in the parasite surface. In addition, we showed that T. cruzi Beclin (TcBeclin1) colocalizes with TcVps34 in epimastigotes, thus suggesting the formation of complexes that may play conserved cellular roles already described for other eukaryotes.
Collapse
Affiliation(s)
- Carlos Alcides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mercedes Soares-Silva
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Y Maeda
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Wanderson Duarte DaRocha
- Laboratório de Genômica Funcional de Parasitos, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Clara Mendes
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Claudio Vieira Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cristina M Orikaza
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Viviane Grazielle Silva
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniella Castanheira Bartholomeu
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
15
|
Reis-Cunha JL, Jeffares DC. Detecting complex infections in trypanosomatids using whole genome sequencing. BMC Genomics 2024; 25:1011. [PMID: 39472783 PMCID: PMC11520695 DOI: 10.1186/s12864-024-10862-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Trypanosomatid parasites are a group of protozoans that cause devastating diseases that disproportionately affect developing countries. These protozoans have developed several mechanisms for adaptation to survive in the mammalian host, such as extensive expansion of multigene families enrolled in host-parasite interaction, adaptation to invade and modulate host cells, and the presence of aneuploidy and polyploidy. Two mechanisms might result in "complex" isolates, with more than two haplotypes being present in a single sample: multiplicity of infections (MOI) and polyploidy. We have developed and validated a methodology to identify multiclonal infections and polyploidy using whole genome sequencing reads, based on fluctuations in allelic read depth in heterozygous positions, which can be easily implemented in experiments sequencing genomes from one sample to larger population surveys. RESULTS The methodology estimates the complexity index (CI) of an isolate, and compares real samples with simulated clonal infections at individual and populational level, excluding regions with somy and gene copy number variation. It was primarily validated with simulated MOI and known polyploid isolates respectively from Leishmania and Trypanosoma cruzi. Then, the approach was used to assess the complexity of infection using genome wide SNP data from 497 trypanosomatid samples from four clades, L. donovani/L. infantum, L. braziliensis, T. cruzi and T. brucei providing an overview of multiclonal infection and polyploidy in these cultured parasites. We show that our method robustly detects complex infections in samples with at least 25x coverage, 100 heterozygous SNPs and where 5-10% of the reads correspond to the secondary clone. We find that relatively small proportions (≤ 7%) of cultured trypanosomatid isolates are complex. CONCLUSIONS The method can accurately identify polyploid isolates, and can identify multiclonal infections in scenarios with sufficient genome read coverage. We pack our method in a single R script that requires only a standard variant call format (VCF) file to run ( https://github.com/jaumlrc/Complex-Infections ). Our analyses indicate that multiclonality and polyploidy do occur in all clades, but not very frequently in cultured trypanosomatids. We caution that our estimates are lower bounds due to the limitations of current laboratory and bioinformatic methods.
Collapse
Affiliation(s)
- João Luís Reis-Cunha
- York Biomedical Research Institute, Department of Biology and York Biomedical Research Institute, University of York, York, YO10 5DD, UK.
| | - Daniel Charlton Jeffares
- York Biomedical Research Institute, Department of Biology and York Biomedical Research Institute, University of York, York, YO10 5DD, UK.
| |
Collapse
|
16
|
Springer AL, Agrawal S, Chang EP. Malate dehydrogenase in parasitic protozoans: roles in metabolism and potential therapeutic applications. Essays Biochem 2024; 68:235-251. [PMID: 38938216 PMCID: PMC11461325 DOI: 10.1042/ebc20230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
The role of malate dehydrogenase (MDH) in the metabolism of various medically significant protozoan parasites is reviewed. MDH is an NADH-dependent oxidoreductase that catalyzes interconversion between oxaloacetate and malate, provides metabolic intermediates for both catabolic and anabolic pathways, and can contribute to NAD+/NADH balance in multiple cellular compartments. MDH is present in nearly all organisms; isoforms of MDH from apicomplexans (Plasmodium falciparum, Toxoplasma gondii, Cryptosporidium spp.), trypanosomatids (Trypanosoma brucei, T. cruzi) and anaerobic protozoans (Trichomonas vaginalis, Giardia duodenalis) are presented here. Many parasitic species have complex life cycles and depend on the environment of their hosts for carbon sources and other nutrients. Metabolic plasticity is crucial to parasite transition between host environments; thus, the regulation of metabolic processes is an important area to explore for therapeutic intervention. Common themes in protozoan parasite metabolism include emphasis on glycolytic catabolism, substrate-level phosphorylation, non-traditional uses of common pathways like tricarboxylic acid cycle and adapted or reduced mitochondria-like organelles. We describe the roles of MDH isoforms in these pathways, discuss unusual structural or functional features of these isoforms relevant to activity or drug targeting, and review current studies exploring the therapeutic potential of MDH and related genes. These studies show that MDH activity has important roles in many metabolic pathways, and thus in the metabolic transitions of protozoan parasites needed for success as pathogens.
Collapse
Affiliation(s)
- Amy L Springer
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, U.S.A
| | - Swati Agrawal
- Department of Biological Sciences, University of Mary Washington, Fredericksburg, VA, U.S.A
| | - Eric P Chang
- Department of Chemistry and Physical Sciences, Pace University, New York, NY, U.S.A
| |
Collapse
|
17
|
Winkler MA, Pan AA. Molecular similarities between the genes for Trypanosoma cruzi microtubule-associated proteins, mammalian interferons, and TRIMs. Parasitol Res 2024; 123:319. [PMID: 39251458 PMCID: PMC11384636 DOI: 10.1007/s00436-024-08329-4] [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: 04/15/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024]
Abstract
Initial studies using bioinformatics analysis revealed DNA sequence similarities between Trypanosoma cruzi GenBank® M21331, coding for Antigen 36 (Ag 36), and tripartite motif (TRIM) genes. TRIM40 showed 9.7% identity to GenBank M21331, and four additional TRIM genes had identities greater than 5.0%. TRIM37 showed a continuous stretch of identity of 12 nucleotides, that is, at least 25% longer than any of the other TRIMs. When we extended our analysis on the relationships of GenBank M21331 to further innate immune genes, using the Needleman-Wunsch (NW) algorithm for alignment, identities to human IFN-α, IFN-β, and IFN-γ genes of 13.6%, 12.6%, and 17.9%, respectively, were found. To determine the minimum number of genes coding for proteins closely related to Ag 36, a BLAST-p search was conducted with it versus the T. cruzi genome. The BLAST-p search revealed that T. cruzi GenBank M21331 had 14 gene sequences homologous to microtubule-associated protein (MAP) genes with 100% amino acid sequence identity. To verify the similarities in non-human genes, a study comparing TRIM21 region sequences among mammalian species to the comparable human TRIM21 region showed that related sequences were also present in 11 mammalian species. The MAP genes homologous to Ag 36 form a family of at least 14 genes which mimic human immune genes in the IFN and TRIM families. This mimicry is of gene sequences and not their protein products or epitopes. These results appear to be the first description of molecular mimicry of immune genes in humans by a protozoan parasite.
Collapse
Affiliation(s)
- Martin A Winkler
- Biotech Advisor, 1321 Wagon Wheel Road, Lawrence, KS, 66049, USA.
| | - Alfred A Pan
- TNTC, Inc, 25A Crescent Drive, Pleasant Hill, CA, 94523, USA
| |
Collapse
|
18
|
Mendes IC, Dos Reis Bertoldo W, Miranda-Junior AS, Assis AVD, Repolês BM, Ferreira WRR, Chame DF, Souza DDL, Pavani RS, Macedo AM, Franco GR, Serra E, Perdomo V, Menck CFM, da Silva Leandro G, Fragoso SP, Barbosa Elias MCQ, Machado CR. DNA lesions that block transcription induce the death of Trypanosoma cruzi via ATR activation, which is dependent on the presence of R-loops. DNA Repair (Amst) 2024; 141:103726. [PMID: 39096697 DOI: 10.1016/j.dnarep.2024.103726] [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: 11/23/2023] [Revised: 06/25/2024] [Accepted: 07/07/2024] [Indexed: 08/05/2024]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease and a peculiar eukaryote with unique biological characteristics. DNA damage can block RNA polymerase, activating transcription-coupled nucleotide excision repair (TC-NER), a DNA repair pathway specialized in lesions that compromise transcription. If transcriptional stress is unresolved, arrested RNA polymerase can activate programmed cell death. Nonetheless, how this parasite modulates these processes is unknown. Here, we demonstrate that T. cruzi cell death after UV irradiation, a genotoxic agent that generates lesions resolved by TC-NER, depends on active transcription and is signaled mainly by an apoptotic-like pathway. Pre-treated parasites with α-amanitin, a selective RNA polymerase II inhibitor, become resistant to such cell death. Similarly, the gamma pre-irradiated cells are more resistant to UV when the transcription processes are absent. The Cockayne Syndrome B protein (CSB) recognizes blocked RNA polymerase and can initiate TC-NER. Curiously, CSB overexpression increases parasites' cell death shortly after UV exposure. On the other hand, at the same time after irradiation, the single-knockout CSB cells show resistance to the same treatment. UV-induced fast death is signalized by the exposition of phosphatidylserine to the outer layer of the membrane, indicating a cell death mainly by an apoptotic-like pathway. Furthermore, such death is suppressed in WT parasites pre-treated with inhibitors of ataxia telangiectasia and Rad3-related (ATR), a key DDR kinase. Signaling for UV radiation death may be related to R-loops since the overexpression of genes associated with the resolution of these structures suppress it. Together, results suggest that transcription blockage triggered by UV radiation activates an ATR-dependent apoptosis-like mechanism in T. cruzi, with the participation of CSB protein in this process.
Collapse
Affiliation(s)
- Isabela Cecilia Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Willian Dos Reis Bertoldo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Adalberto Sales Miranda-Junior
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Antônio Vinícius de Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Bruno Marçal Repolês
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Wesley Roger Rodrigues Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela Ferreira Chame
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela De Laet Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Raphael Souza Pavani
- Laboratório Especial de Ciclo Celular, Instituto Butantan, São Paulo, MG, São Paulo, SP 05503-900, Brazil
| | - Andrea Mara Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Glória Regina Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Esteban Serra
- Instituto de Biología Molecular y Celular de Rosario, CONICET, 2000 Rosario, Santa Fe, Argentina; Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Virginia Perdomo
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Carlos Frederico Martins Menck
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | - Giovana da Silva Leandro
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | | | | | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil.
| |
Collapse
|
19
|
Bombaça ACS, Caminha MA, Barbosa JMC, Pedra-Rezende Y, Ennes-Vidal V, Brunoro GVF, Archanjo BS, d'Avila CM, Valente RH, Menna-Barreto RFS. Heme metabolism in Strigomonas culicis: Implications of H 2O 2 resistance induction and symbiont elimination. J Biol Chem 2024; 300:107692. [PMID: 39159809 PMCID: PMC11648444 DOI: 10.1016/j.jbc.2024.107692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
Monoxenous trypanosomatid Strigomonas culicis harbors an endosymbiotic bacterium, which enables the protozoa to survive without heme supplementation. The impact of H2O2 resistance and symbiont elimination on intracellular heme and Fe2+ availability was analyzed through a comparison of WT strain with both WT H2O2-resistant (WTR) and aposymbiotic (Apo) protozoa. The relative quantification of the heme biosynthetic pathway through label-free parallel reaction monitoring targeted mass spectrometry revealed that H2O2 resistance does not influence the abundance of tryptic peptides. However, the Apo strain showed increased coproporphyrinogen III oxidase and ferrochelatase levels. A putative ferrous iron transporter, homologous to LIT1 and TcIT from Leishmania major and Trypanosoma cruzi, was identified for the first time. Label-free parallel reaction monitoring targeted mass spectrometry also showed that S. culicis Iron Transporter (ScIT) increased 1.6- and 16.4-fold in WTR and Apo strains compared to WT. Accordingly, antibody-mediated blockage of ScIT decreased by 28.0% and 40.0% intracellular Fe2+concentration in both WTR and Apo strains, whereas no effect was detected in WT. In a heme-depleted medium, adding 10 μM hemin decreased ScIT transcript levels in Apo, whereas 10 μM PPIX, the substrate of ferrochelatase, increased intracellular Fe2+ concentration and ferric iron reduction. Overall, the data suggest mechanisms dependent on de novo heme synthesis (and its substrates) in the Apo strain to overcome reduced heme availability. Given the importance of heme and Fe2+ as cofactors in metabolic pathways, including oxidative phosphorylation and antioxidant systems, this study provides novel mechanistic insights associated with H2O2 resistance in S. culicis.
Collapse
Affiliation(s)
- Ana Cristina Souza Bombaça
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marcelle Almeida Caminha
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | | | - Yasmin Pedra-Rezende
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | - Vitor Ennes-Vidal
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Bráulio Soares Archanjo
- Divisão de Metrologia de Materiais, Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Duque de Caxias, Brazil
| | - Claudia Masini d'Avila
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Richard Hemmi Valente
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | | |
Collapse
|
20
|
Barnadas-Carceller B, Del Portillo HA, Fernandez-Becerra C. Extracellular vesicles as biomarkers in parasitic disease diagnosis. CURRENT TOPICS IN MEMBRANES 2024; 94:187-223. [PMID: 39370207 DOI: 10.1016/bs.ctm.2024.07.003] [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: 10/08/2024]
Abstract
Parasitic diseases constitute a major global health problem, affecting millions of people worldwide. Recent advances in the study of extracellular vesicles (EVs) have opened up new strategies for biomarker discovery in protozoan and helminth infections. Analyses of EVs in cultures and biological fluids have identified numerous potential biomarkers that could be useful for early and differential diagnosis, monitoring therapeutic responses, and the overall management and control of these diseases. Despite the potential of these biomarkers, several challenges must be addressed, including limited research, the need for standardized protocols, and the reproducibility of results across studies. In many parasitic infections, EVs have been obtained from various sample types, including plasma from human patients and mouse models, as well as cultures of the parasites at different stages. EVs were isolated by various methods and predominantly characterized through proteomic analysis or RNA sequencing to assess their cargo and identify potential biomarkers. These biomarker candidates were investigated and validated using different assays such as ELISA, Western Blot, and ROC curves. Overall, the use of EVs is considered a promising new diagnostic strategy for parasite infections, but further research with larger cohorts, standardized methods, and additional validation tests are essential for effective diagnosis and management of these diseases.
Collapse
Affiliation(s)
- Berta Barnadas-Carceller
- ISGlobal, Barcelona Institute for Global Health, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; IGTP Institut d'Investigació Germans Trias I Pujol, Badalona, Barcelona, Spain
| | - Hernando A Del Portillo
- ISGlobal, Barcelona Institute for Global Health, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; IGTP Institut d'Investigació Germans Trias I Pujol, Badalona, Barcelona, Spain; ICREA, Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Carmen Fernandez-Becerra
- ISGlobal, Barcelona Institute for Global Health, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; IGTP Institut d'Investigació Germans Trias I Pujol, Badalona, Barcelona, Spain; CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
21
|
Kieft R, Reynolds D, Sabatini R. Epigenetic regulation of TERRA transcription and metacyclogenesis by base J in Leishmania major. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601056. [PMID: 38979290 PMCID: PMC11230386 DOI: 10.1101/2024.06.27.601056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The hyper-modified DNA base J helps control termination of Pol II transcription at polycistronic transcription units (PTUs) in T. brucei and L. major , allowing epigenetic control of gene expression. The Telomere Repeat-containing RNA (TERRA) is synthesized in T. brucei by Pol I readthrough transcription of a telomeric PTU. While little is understood regarding TERRA synthesis and function, the hyper-modified DNA base J is highly enriched at telomeres in L. major promastigotes. We now show that TERRA is synthesized by Pol II in L. major and loss of base J leads to increased TERRA. For at least one site, the increased TERRA is by Pol II readthrough transcription from an adjacent PTU. Furthermore, Pol II readthrough defects and increased TERRA correlate with increased differentiation of promastigotes to the infectious metacyclic life stage and decreased cell viability. These results help explain the essential nature of base J in Leishmania and provide insight regarding epigenetic control of coding and non-coding RNA expression and parasite development during the life cycle of L. major .
Collapse
|
22
|
Hakim JMC, Gutierrez Guarnizo SA, Málaga Machaca E, Gilman RH, Mugnier MR. Whole-genome assembly of a hybrid Trypanosoma cruzi strain assembled with Nanopore sequencing alone. G3 (BETHESDA, MD.) 2024; 14:jkae076. [PMID: 38592968 PMCID: PMC11152063 DOI: 10.1093/g3journal/jkae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/12/2023] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, which causes 10,000 deaths per year. Despite the high mortality associated with Chagas, relatively few parasite genomes have been assembled to date, with genome assemblies unavailable even for some commonly used laboratory strains. This is at least partially due to T. cruzi's highly complex and highly repetitive genome, which defies investigation using traditional short-read sequencing methods. In this study, we have generated a high-quality whole-genome assembly of the hybrid Tulahuen strain, a commercially available type VI strain, using long-read Nanopore sequencing without short-read scaffolding. The assembled genome contains 25% repeat regions, 17% variable multigene family members, and 27% transposable elements (TEs) and is of comparable quality with T. cruzi genome assemblies that utilized both long- and short-read data. Notably, we find that regions with TEs are significantly enriched for multicopy surface proteins, and that surface proteins are, on average, closer to TEs than to other coding regions. This finding suggests that mobile genetic elements such as transposons may drive recombination within surface protein gene families. This work demonstrates the feasibility of Nanopore sequencing to resolve complex regions of T. cruzi genomes, and with these resolved regions, provides support for a possible mechanism for genomic diversification.
Collapse
Affiliation(s)
- Jill M C Hakim
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Edith Málaga Machaca
- Asociación Benéfica PRISMA, Lima 15102, Peru
- Infectious Diseases Research Laboratory, Department of Cellular and Molecular Sciences, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Robert H Gilman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Monica R Mugnier
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
23
|
Cárdenas-Guerra RE, Montes-Flores O, Nava-Pintor EE, Reséndiz-Cardiel G, Flores-Pucheta CI, Rodríguez-Gavaldón YI, Arroyo R, Bottazzi ME, Hotez PJ, Ortega-López J. Chagasin from Trypanosoma cruzi as a molecular scaffold to express epitopes of TSA-1 as soluble recombinant chimeras. Protein Expr Purif 2024; 218:106458. [PMID: 38423156 DOI: 10.1016/j.pep.2024.106458] [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: 09/05/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, a global public health problem. New therapeutic drugs and biologics are needed. The TSA-1 recombinant protein of T. cruzi is one such promising antigen for developing a therapeutic vaccine. However, it is overexpressed in E. coli as inclusion bodies, requiring an additional refolding step. As an alternative, in this study, we propose the endogenous cysteine protease inhibitor chagasin as a molecular scaffold to generate chimeric proteins. These proteins will contain combinations of two of the five conserved epitopes (E1 to E5) of TSA-1 in the L4 and L6 chagasin loops. Twenty chimeras (Q1-Q20) were designed, and their solubility was predicted using bioinformatics tools. Nine chimeras with different degrees of solubility were selected and expressed in E. coli BL21 (DE3). Western blot assays with anti-6x-His and anti-chagasin antibodies confirmed the expression of soluble recombinant chimeras. Both theoretically and experimentally, the Q12 (E5-E3) chimera was the most soluble, and the Q20 (E4-E5) the most insoluble protein. Q4 (E5-E1) and Q8 (E5-E2) chimeras were classified as proteins with medium solubility that exhibited the highest yield in the soluble fraction. Notably, Q4 has a yield of 239 mg/L, well above the yield of recombinant chagasin (16.5 mg/L) expressed in a soluble form. The expression of the Q4 chimera was scaled up to a 7 L fermenter obtaining a yield of 490 mg/L. These data show that chagasin can serve as a molecular scaffold for the expression of TSA-1 epitopes in the form of soluble chimeras.
Collapse
Affiliation(s)
- Rosa Elena Cárdenas-Guerra
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Octavio Montes-Flores
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Edgar Ezequiel Nava-Pintor
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Gerardo Reséndiz-Cardiel
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Claudia Ivonne Flores-Pucheta
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Yasmín Irene Rodríguez-Gavaldón
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jaime Ortega-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico.
| |
Collapse
|
24
|
Alonaizan R. Molecular regulation of NLRP3 inflammasome activation during parasitic infection. Biosci Rep 2024; 44:BSR20231918. [PMID: 38623843 PMCID: PMC11096646 DOI: 10.1042/bsr20231918] [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: 11/05/2023] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024] Open
Abstract
Parasitic diseases are a serious global health concern, causing many common and severe infections, including Chagas disease, leishmaniasis, and schistosomiasis. The NLRP3 inflammasome belongs to the NLR (nucleotide-binding domain leucine-rich-repeat-containing proteins) family, which are cytosolic proteins playing key roles in the detection of pathogens. NLRP3 inflammasomes are activated in immune responses to Plasmodium, Leishmania, Toxoplasma gondii, Entamoeba histolytica, Trypanosoma cruzi, and other parasites. The role of NLRP3 is not fully understood, but it is a crucial component of the innate immune response to parasitic infections and its functions as a sensor triggering the inflammatory response to the invasive parasites. However, while this response can limit the parasites' growth, it can also result in potentially catastrophic host pathology. This makes it essential to understand how NLRP3 interacts with parasites to initiate the inflammatory response. Plasmodium hemozoin, Leishmania glycoconjugate lipophosphoglycan (LPG) and E. histolytica Gal/GalNAc lectin can stimulate NLRP3 activation, while the dense granule protein 9 (GRA9) of T. gondii has been shown to suppress it. Several other parasitic products also have diverse effects on NLRP3 activation. Understanding the mechanism of NLRP3 interaction with these products will help to develop advanced therapeutic approaches to treat parasitic diseases. This review summarizes current knowledge of the NLRP3 inflammasome's action on the immune response to parasitic infections and aims to determine the mechanisms through which parasitic molecules either activate or inhibit its action.
Collapse
Affiliation(s)
- Rasha Alonaizan
- Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
25
|
Marinov GK, Chen X, Swaffer MP, Xiang T, Grossman AR, Greenleaf WJ. Genome-wide distribution of 5-hydroxymethyluracil and chromatin accessibility in the Breviolum minutum genome. Genome Biol 2024; 25:115. [PMID: 38711126 PMCID: PMC11071213 DOI: 10.1186/s13059-024-03261-3] [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: 09/23/2023] [Accepted: 04/28/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND In dinoflagellates, a unique and extremely divergent genomic and nuclear organization has evolved. The highly unusual features of dinoflagellate nuclei and genomes include permanently condensed liquid crystalline chromosomes, primarily packaged by proteins other than histones, genes organized in very long unidirectional gene arrays, a general absence of transcriptional regulation, high abundance of the otherwise very rare DNA modification 5-hydroxymethyluracil (5-hmU), and many others. While most of these fascinating properties are originally identified in the 1970s and 1980s, they have not yet been investigated using modern genomic tools. RESULTS In this work, we address some of the outstanding questions regarding dinoflagellate genome organization by mapping the genome-wide distribution of 5-hmU (using both immunoprecipitation-based and basepair-resolution chemical mapping approaches) and of chromatin accessibility in the genome of the Symbiodiniaceae dinoflagellate Breviolum minutum. We find that the 5-hmU modification is preferentially enriched over certain classes of repetitive elements, often coincides with the boundaries between gene arrays, and is generally correlated with decreased chromatin accessibility, the latter otherwise being largely uniform along the genome. We discuss the potential roles of 5-hmU in the functional organization of dinoflagellate genomes and its relationship to the transcriptional landscape of gene arrays. CONCLUSIONS Our results provide the first window into the 5-hmU and chromatin accessibility landscapes in dinoflagellates.
Collapse
Affiliation(s)
- Georgi K Marinov
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA.
| | - Xinyi Chen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Matthew P Swaffer
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Tingting Xiang
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, 94305, USA
| | - Arthur R Grossman
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, 94305, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA.
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, 94305, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, 94305, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
26
|
Vitarelli MDO, Franco TA, Pires DDS, Lima ARJ, Viala VL, Kraus AJ, de Azevedo IDLMJ, da Cunha JPC, Elias MC. Integrating high-throughput analysis to create an atlas of replication origins in Trypanosoma cruzi in the context of genome structure and variability. mBio 2024; 15:e0031924. [PMID: 38441981 PMCID: PMC11005370 DOI: 10.1128/mbio.00319-24] [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: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
Trypanosoma cruzi is the etiologic agent of the most prevalent human parasitic disease in Latin America, Chagas disease. Its genome is rich in multigenic families that code for virulent antigens and are present in the rapidly evolving genomic compartment named Disruptive. DNA replication is a meticulous biological process in which flaws can generate mutations and changes in chromosomal and gene copy numbers. Here, integrating high-throughput and single-molecule analyses, we were able to identify Predominant, Flexible, and Dormant Orc1Cdc6-dependent origins as well as Orc1Cdc6-independent origins. Orc1Cdc6-dependent origins were found in multigenic family loci, while independent origins were found in the Core compartment that contains conserved and hypothetical protein-coding genes, in addition to multigenic families. In addition, we found that Orc1Cdc6 density is related to the firing of origins and that Orc1Cdc6-binding sites within fired origins are depleted of a specific class of nucleosomes that we previously categorized as dynamic. Together, these data suggest that Orc1Cdc6-dependent origins may contribute to the rapid evolution of the Disruptive compartment and, therefore, to the success of T. cruzi infection and that the local epigenome landscape is also involved in this process.IMPORTANCETrypanosoma cruzi, responsible for Chagas disease, affects millions globally, particularly in Latin America. Lack of vaccine or treatment underscores the need for research. Parasite's genome, with virulent antigen-coding multigenic families, resides in the rapidly evolving Disruptive compartment. Study sheds light on the parasite's dynamic DNA replication, discussing the evolution of the Disruptive compartment. Therefore, the findings represent a significant stride in comprehending T. cruzi's biology and the molecular bases that contribute to the success of infection caused by this parasite.
Collapse
Affiliation(s)
- Marcela de Oliveira Vitarelli
- Cell Cycle Laboratory, Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
| | | | | | | | - Vincent Louis Viala
- Biochemistry Laboratory, Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
| | - Amelie Johanna Kraus
- Division of Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried, Germany
- Biomedical Center, Division of Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried, Germany
| | | | - Julia Pinheiro Chagas da Cunha
- Cell Cycle Laboratory, Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
| | - Maria Carolina Elias
- Cell Cycle Laboratory, Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brazil, São Paulo, Brazil
| |
Collapse
|
27
|
Rossi IV, de Souza DAS, Ramirez MI. The End Justifies the Means: Chagas Disease from a Perspective of the Host- Trypanosoma cruzi Interaction. Life (Basel) 2024; 14:488. [PMID: 38672758 PMCID: PMC11050810 DOI: 10.3390/life14040488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
The neglected Chagas disease (CD) is caused by the protozoan parasite Trypanosoma cruzi. Despite CD dispersion throughout the world, it prevails in tropical areas affecting mainly poor communities, causing devastating health, social and economic consequences. Clinically, CD is marked by a mildly symptomatic acute phase, and a chronic phase characterized by cardiac and/or digestive complications. Current treatment for CD relies on medications with strong side effects and reduced effectiveness. The complex interaction between the parasite and the host outlines the etiology and progression of CD. The unique characteristics and high adaptability of T. cruzi, its mechanisms of persistence, and evasion of the immune system seem to influence the course of the disease. Despite the efforts to uncover the pathology of CD, there are many gaps in understanding how it is established and reaches chronicity. Also, the lack of effective treatments and protective vaccines constitute challenges for public health. Here, we explain the background in which CD is established, from the peculiarities of T. cruzi molecular biology to the development of the host's immune response leading to the pathophysiology of CD. We also discuss the state of the art of treatments for CD and current challenges in basic and applied science.
Collapse
Affiliation(s)
- Izadora Volpato Rossi
- Graduate Program in Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| | - Denise Andréa Silva de Souza
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| | - Marcel Ivan Ramirez
- Graduate Program in Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| |
Collapse
|
28
|
Romagnoli BAA, Lucena ACR, Freire ER, Munhoz da Rocha IF, Alves LR, Goldenberg S. TcZC3HTTP, a regulatory element that contributes to Trypanosoma cruzi cell proliferation. Microbiol Spectr 2024; 12:e0288023. [PMID: 38270449 PMCID: PMC10913370 DOI: 10.1128/spectrum.02880-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024] Open
Abstract
Post-transcriptional regulation of gene expression is a critical process for adapting to and surviving Trypanosoma cruzi, a parasite with a complex life cycle. RNA-binding proteins (RBPs) are key players in this regulation, forming ribonucleoprotein complexes (messenger ribonucleoproteins) and RNA granules that control transcript stability, localization, degradation, and translation modulation. Understanding the specific roles of individual RBPs is crucial for unraveling the details of this regulatory network. In this study, we generated null mutants of the TcZC3HTTP gene, a specific RBP in the Trypanosoma family characterized by a C3H zinc finger and a DNAJ domain associated with RNA and protein binding, respectively. Through cell growth assays, we demonstrated that the absence of TcZC3HTTP or the expression of an additional tagged version impacted epimastigote growth, indicating its contribution to cell proliferation. TcZC3HTTP was found to associate with mRNAs involved in cell cycle and division in epimastigotes, while in nutritionally stressed parasites it exhibited associations with mRNAs coding for other RBPs and rRNA. Furthermore, our analysis identified that TcZC3HTTP protein partners were different during normal growth conditions compared to starvation conditions, with the latter showing enrichment of ribosomal proteins and other RBPs. Therefore, this study provides insights into TcZC3HTTP's role in the post-transcriptional regulation of gene expression during normal growth and nutritional stress in T. cruzi, uncovering its versatile functions in different cellular contexts.IMPORTANCEUnderstanding how Trypanosoma cruzi, the causative agent of Chagas disease, regulates gene expression is crucial for developing targeted interventions. In this study, we investigated the role of TcZC3HTTP, an RNA-binding protein, in post-transcriptional regulation. Our findings demonstrate that TcZC3HTTP is relevant for the growth and proliferation of epimastigotes, a stage of the parasite's life cycle. We identified its associations with specific mRNAs involved in cell cycle and division and its interactions with enzymes and other RNA-binding proteins (RBPs) under normal and starvation conditions. These insights shed light on the regulatory network underlying gene expression in T. cruzi and reveal the multifaceted functions of RBPs in this parasite. Such knowledge enhances our understanding of the parasite's biology and opens avenues for developing novel therapeutic strategies targeting post-transcriptional gene regulation in T. cruzi.
Collapse
Affiliation(s)
| | - Aline Castro Rodrigues Lucena
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Fiocruz PR, Curitiba, Paraná, Brazil
| | - Eden Ribeiro Freire
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, Fiocruz PR, Curitiba, Paraná, Brazil
| | | | - Lysangela Ronalte. Alves
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, Fiocruz PR, Curitiba, Paraná, Brazil
- Research Center in Infectious Diseases, Division of Infectious Disease and Immunity, CHU de Quebec Research Center, University Laval, Quebec, Canada
| | - Samuel Goldenberg
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, Fiocruz PR, Curitiba, Paraná, Brazil
| |
Collapse
|
29
|
Matos ÂP, Saldanha-Corrêa FMP, Gomes RDS, Hurtado GR. Exploring microalgal and cyanobacterial metabolites with antiprotozoal activity against Leishmania and Trypanosoma parasites. Acta Trop 2024; 251:107116. [PMID: 38159713 DOI: 10.1016/j.actatropica.2023.107116] [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: 10/25/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Neglected tropical diseases (NTD) like Leishmaniasis and trypanosomiasis affect millions of people annually, while currently used antiprotozoal drugs have serious side effects. Drug research based on natural products has shown that microalgae and cyanobacteria are a promising platform of biochemically active compounds with antiprotozoal activity. These unicellular photosynthetic organisms are rich in polyunsaturated fatty acids, pigments including phycocyanin, chlorophylls and carotenoids, polyphenols, bioactive peptides, terpenes, alkaloids, which have proven antioxidant, antimicrobial, antiviral, antiplasmodial and antiprotozoal properties. This review provides up-to-date information regarding ongoing studies on substances synthesized by microalgae and cyanobacteria with notable activity against Leishmania spp., Trypanosoma cruzi, and Trypanosoma brucei, the causative agents of Leishmaniasis, Chagas disease, and human African trypanosomiasis, respectively. Extracts of several freshwater or marine microalgae have been tested on different strains of Leishmania and Trypanosoma parasites. For instance, ethanolic extract of Chlamydomonas reinhardtii and Tetraselmis suecica have biological activity against T. cruzi, due to their high content of carotenoids, chlorophylls, phenolic compounds and flavonoids that are associated with trypanocidal activity. Halophilic Dunaliella salina showed moderate antileishmanial activity that may be attributed to the high β-carotene content in this microalga. Peptides such as almiramides, dragonamides, and herbamide that are biosynthesized by marine cyanobacteria Lyngbya majuscula were found to have increased activity in micromolar scale IC50 against L. donovani, T. Cruzi, and T. brucei parasites. The cyanobacterial peptides symplocamide and venturamide isolated from Symploca and Oscillatoria species, respectively, and the alkaloid nostocarbonile isolated from Nostoc have shown promising antiprotozoal properties and are being explored for pharmaceutical and medicinal purposes. The discovery of new molecules from microalgae and cyanobacteria with therapeutic potential against Leishmaniasis and trypanosomiasis may address an urgent medical need: effective and safe treatments of NTDs.
Collapse
Affiliation(s)
- Ângelo Paggi Matos
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil.
| | | | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Gabriela Ramos Hurtado
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil; Institute of Science and Technology, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil.
| |
Collapse
|
30
|
Dadzie FA, Beaudry MS, Deyanov A, Slanis H, Duong MQ, Turner R, Khan A, Arias CA, Kissinger JC, Glenn TC, de Paula Baptista R. "Evaluating the Benefits and Limits of Multiple Displacement Amplification with Whole-Genome Oxford Nanopore Sequencing". BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579537. [PMID: 38405857 PMCID: PMC10888933 DOI: 10.1101/2024.02.09.579537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Multiple Displacement Amplification (MDA) outperforms conventional PCR in long fragment and whole genome amplification which makes it attractive to couple with long-read sequencing of samples with limited quantities of DNA to obtain improved genome assemblies. Here, we explore the efficacy and limits of MDA for genome sequence assembly using Oxford Nanopore Technologies (ONT) rapid library preparations and minION sequencing. We successfully generated almost complete genome sequences for all organisms examined, including Cryptosporidium meleagridis, Staphylococcus aureus, Enterococcus faecium, and Escherichia coli, with the ability to generate high-quality data from samples starting with only 0.025 ng of total DNA. Controlled sheared DNA samples exhibited a distinct pattern of size-increase after MDA, which may be associated with the amplification of long, low-abundance fragments present in the assay, as well as generating concatemeric sequences during amplification. To address concatemers, we developed a computational pipeline (CADECT: Concatemer Detection Tool) to identify and remove putative concatemeric sequences. This study highlights the efficacy of MDA in generating high-quality genome assemblies from limited amounts of input DNA. Also, the CADECT pipeline effectively mitigated the impact of concatemeric sequences, enabling the assembly of contiguous sequences even in cases where the input genomic DNA was degraded. These results have significant implications for the study of organisms that are challenging to culture in vitro, such as Cryptosporidium, and for expediting critical results in clinical settings with limited quantities of available genomic DNA.
Collapse
Affiliation(s)
- Fiifi A. Dadzie
- Department of Genetics, University of Georgia, Athens, GA USA 30602
| | - Megan S. Beaudry
- Department of Environmental Health Science, University of Georgia, Athens, GA USA 30602
| | - Alex Deyanov
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX USA 77030
| | - Haley Slanis
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX USA 77030
| | - Minh Q. Duong
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX USA 77030
| | - Randi Turner
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA USA
- USA Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Service, Animal Parasitic Disease Laboratory, Beltsville, MD USA
| | - Asis Khan
- USA Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Service, Animal Parasitic Disease Laboratory, Beltsville, MD USA
| | - Cesar A. Arias
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX USA 77030
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, TX USA 77030
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jessica C. Kissinger
- Department of Genetics, University of Georgia, Athens, GA USA 30602
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA USA
- Institute of Bioinformatics, University of Georgia, Athens, GA USA 30602
| | - Travis C. Glenn
- Department of Genetics, University of Georgia, Athens, GA USA 30602
- Department of Environmental Health Science, University of Georgia, Athens, GA USA 30602
- Institute of Bioinformatics, University of Georgia, Athens, GA USA 30602
| | - Rodrigo de Paula Baptista
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX USA 77030
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA USA
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, TX USA 77030
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Institute of Bioinformatics, University of Georgia, Athens, GA USA 30602
| |
Collapse
|
31
|
Hoyos Sanchez MC, Ospina Zapata HS, Suarez BD, Ospina C, Barbosa HJ, Carranza Martinez JC, Vallejo GA, Urrea Montes D, Duitama J. A phased genome assembly of a Colombian Trypanosoma cruzi TcI strain and the evolution of gene families. Sci Rep 2024; 14:2054. [PMID: 38267502 PMCID: PMC10808112 DOI: 10.1038/s41598-024-52449-x] [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: 08/03/2023] [Accepted: 01/18/2024] [Indexed: 01/26/2024] Open
Abstract
Chagas is an endemic disease in tropical regions of Latin America, caused by the parasite Trypanosoma cruzi. High intraspecies variability and genome complexity have been challenges to assemble high quality genomes needed for studies in evolution, population genomics, diagnosis and drug development. Here we present a chromosome-level phased assembly of a TcI T. cruzi strain (Dm25). While 29 chromosomes show a large collinearity with the assembly of the Brazil A4 strain, three chromosomes show both large heterozygosity and large divergence, compared to previous assemblies of TcI T. cruzi strains. Nucleotide and protein evolution statistics indicate that T. cruzi Marinkellei separated before the diversification of T. cruzi in the known DTUs. Interchromosomal paralogs of dispersed gene families and histones appeared before but at the same time have a more strict purifying selection, compared to other repeat families. Previously unreported large tandem arrays of protein kinases and histones were identified in this assembly. Over one million variants obtained from Illumina reads aligned to the primary assembly clearly separate the main DTUs. We expect that this new assembly will be a valuable resource for further studies on evolution and functional genomics of Trypanosomatids.
Collapse
Affiliation(s)
- Maria Camila Hoyos Sanchez
- Systems and Computing Engineering Department, Universidad de los Andes, Bogotá, Colombia
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, 79106, USA
| | | | - Brayhan Dario Suarez
- Laboratorio de Investigaciones en Parasitología Tropical (LIPT), Universidad del Tolima, Ibagué, Colombia
| | - Carlos Ospina
- Laboratorio de Investigaciones en Parasitología Tropical (LIPT), Universidad del Tolima, Ibagué, Colombia
| | - Hamilton Julian Barbosa
- Laboratorio de Investigaciones en Parasitología Tropical (LIPT), Universidad del Tolima, Ibagué, Colombia
| | | | - Gustavo Adolfo Vallejo
- Laboratorio de Investigaciones en Parasitología Tropical (LIPT), Universidad del Tolima, Ibagué, Colombia
| | - Daniel Urrea Montes
- Laboratorio de Investigaciones en Parasitología Tropical (LIPT), Universidad del Tolima, Ibagué, Colombia
| | - Jorge Duitama
- Systems and Computing Engineering Department, Universidad de los Andes, Bogotá, Colombia.
| |
Collapse
|
32
|
Ossowski MS, Gallardo JP, Niborski LL, Rodríguez-Durán J, Lapadula WJ, Juri Ayub M, Chadi R, Hernandez Y, Fernandez ML, Potenza M, Gómez KA. Characterization of Novel Trypanosoma cruzi-Specific Antigen with Potential Use in the Diagnosis of Chagas Disease. Int J Mol Sci 2024; 25:1202. [PMID: 38256275 PMCID: PMC10816184 DOI: 10.3390/ijms25021202] [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: 11/07/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Chagas disease is caused by the parasite Trypanosoma cruzi. In humans, it evolves into a chronic disease, eventually resulting in cardiac, digestive, and/or neurological disorders. In the present study, we characterized a novel T. cruzi antigen named Tc323 (TcCLB.504087.20), recognized by a single-chain monoclonal antibody (scFv 6B6) isolated from the B cells of patients with cardiomyopathy related to chronic Chagas disease. Tc323, a ~323 kDa protein, is an uncharacterized protein showing putative quinoprotein alcohol dehydrogenase-like domains. A computational molecular docking study revealed that the scFv 6B6 binds to an internal domain of Tc323. Immunofluorescence microscopy and Western Blot showed that Tc323 is expressed in the main developmental forms of T. cruzi, localized intracellularly and exhibiting a membrane-associated pattern. According to phylogenetic analysis, Tc323 is highly conserved throughout evolution in all the lineages of T. cruzi so far identified, but it is absent in Leishmania spp. and Trypanosoma brucei. Most interestingly, only plasma samples from patients infected with T. cruzi and those with mixed infection with Leishmania spp. reacted against Tc323. Collectively, our findings demonstrate that Tc323 is a promising candidate for the differential serodiagnosis of chronic Chagas disease in areas where T. cruzi and Leishmania spp. infections coexist.
Collapse
Affiliation(s)
- Micaela S. Ossowski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Juan Pablo Gallardo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Leticia L. Niborski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Jessica Rodríguez-Durán
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Walter J. Lapadula
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), Facultad de Química Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis 5700, Argentina; (W.J.L.); (M.J.A.)
| | - Maximiliano Juri Ayub
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), Facultad de Química Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis 5700, Argentina; (W.J.L.); (M.J.A.)
| | - Raúl Chadi
- Hospital General de Agudos “Dr. Ignacio Pirovano”, Buenos Aires 1430, Argentina;
| | - Yolanda Hernandez
- Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Buenos Aires 1063, Argentina; (Y.H.); (M.L.F.)
| | - Marisa L. Fernandez
- Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Buenos Aires 1063, Argentina; (Y.H.); (M.L.F.)
| | - Mariana Potenza
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Karina A. Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| |
Collapse
|
33
|
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: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
34
|
Tullume-Vergara PO, Caicedo KYO, Tantalean JFC, Serrano MG, Buck GA, Teixeira MMG, Shaw JJ, Alves JMP. Genomes of Endotrypanum monterogeii from Panama and Zelonia costaricensis from Brazil: Expansion of Multigene Families in Leishmaniinae Parasites That Are Close Relatives of Leishmania spp. Pathogens 2023; 12:1409. [PMID: 38133293 PMCID: PMC10747355 DOI: 10.3390/pathogens12121409] [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: 10/23/2023] [Revised: 11/10/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
The Leishmaniinae subfamily of the Trypanosomatidae contains both genus Zelonia (monoxenous) and Endotrypanum (dixenous). They are amongst the nearest known relatives of Leishmania, which comprises many human pathogens widespread in the developing world. These closely related lineages are models for the genomic biology of monoxenous and dixenous parasites. Herein, we used comparative genomics to identify the orthologous groups (OGs) shared among 26 Leishmaniinae species to investigate gene family expansion/contraction and applied two phylogenomic approaches to confirm relationships within the subfamily. The Endotrypanum monterogeii and Zelonia costaricensis genomes were assembled, with sizes of 29.9 Mb and 38.0 Mb and 9.711 and 12.201 predicted protein-coding genes, respectively. The genome of E. monterogeii displayed a higher number of multicopy cell surface protein families, including glycoprotein 63 and glycoprotein 46, compared to Leishmania spp. The genome of Z. costaricensis presents expansions of BT1 and amino acid transporters and proteins containing leucine-rich repeat domains, as well as a loss of ABC-type transporters. In total, 415 and 85 lineage-specific OGs were identified in Z. costaricensis and E. monterogeii. The evolutionary relationships within the subfamily were confirmed using the supermatrix (3384 protein-coding genes) and supertree methods. Overall, this study showed new expansions of multigene families in monoxenous and dixenous parasites of the subfamily Leishmaniinae.
Collapse
Affiliation(s)
- Percy O. Tullume-Vergara
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| | - Kelly Y. O. Caicedo
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| | - Jose F. C. Tantalean
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| | - Myrna G. Serrano
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, 1101 E Marshall St., Richmond, VA 23298, USA; (M.G.S.); (G.A.B.)
| | - Gregory A. Buck
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, 1101 E Marshall St., Richmond, VA 23298, USA; (M.G.S.); (G.A.B.)
| | - Marta M. G. Teixeira
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| | - Jeffrey J. Shaw
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| | - Joao M. P. Alves
- Department of Parasitology, Institute for Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, Sao Paulo 05508-000, SP, Brazil; (P.O.T.-V.); (K.Y.O.C.); (J.F.C.T.); (M.M.G.T.); (J.J.S.)
| |
Collapse
|
35
|
Beati P, Massimino Stepñicka M, Vilchez Larrea SC, Smircich P, Alonso GD, Ocampo J. Improving genome-wide mapping of nucleosomes in Trypanosome cruzi. PLoS One 2023; 18:e0293809. [PMID: 37988351 PMCID: PMC10662739 DOI: 10.1371/journal.pone.0293809] [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: 04/04/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023] Open
Abstract
In Trypanosoma cruzi DNA is packaged into chromatin by octamers of histone proteins that form nucleosomes. Transcription of protein coding genes in trypanosomes is constitutive producing polycistronic units and gene expression is primarily regulated post-transcriptionally. However, chromatin organization influences DNA dependent processes. Hence, determining nucleosome position is of uppermost importance to understand the peculiarities found in trypanosomes. To map nucleosomes genome-wide in several organisms, digestion of chromatin with micrococcal nuclease followed by deep sequencing has been applied. Nonetheless, the special requirements for cell manipulation and the uniqueness of the chromatin organization in trypanosomes entails a customized analytical approach. In this work, we adjusted this broadly used method to the hybrid reference strain, CL Brener. Particularly, we implemented an exhaustive and thorough computational workflow to overcome the difficulties imposed by this complex genome. We tested the performance of two aligners, Bowtie2 and HISAT2, and discuss their advantages and caveats. Specifically, we highlight the relevance of using the whole genome as a reference instead of the commonly used Esmeraldo-like haplotype to avoid spurious alignments. Additionally, we show that using the whole genome refines the average nucleosome representation, but also the quality of mapping for every region represented. Moreover, we show that the average nucleosome organization around trans-splicing acceptor site described before, is not just an average since the same chromatin pattern is detected for most of the represented regions. In addition, we extended the study to a non-hybrid strain applying the experimental and analytical approach to Sylvio-X10 strain. Furthermore, we provide a source code for the construction of 2D plots and heatmaps which are easy to adapt to any T. cruzi strain.
Collapse
Affiliation(s)
- Paula Beati
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Milena Massimino Stepñicka
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Salomé C. Vilchez Larrea
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Smircich
- Laboratorio de Bioinformática, Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Guillermo D. Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Josefina Ocampo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
36
|
Silva Dias Vieira C, Pinheiro Aguiar R, de Almeida Nogueira NP, Costa dos Santos Junior G, Paes MC. Glucose metabolism sustains heme-induced Trypanosoma cruzi epimastigote growth in vitro. PLoS Negl Trop Dis 2023; 17:e0011725. [PMID: 37948458 PMCID: PMC10664871 DOI: 10.1371/journal.pntd.0011725] [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: 06/05/2023] [Revised: 11/22/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
Chagas disease is caused by the protozoan parasite, Trypanosoma cruzi. This parasite alternates between an insect vector and a mammalian host. T. cruzi epimastigotes reside in the insect vector and coexist with the blood components of the vertebrate host. The metabolic profile of T. cruzi has been extensively studied; however, changes in its metabolism in response to signaling molecules present in the vector are poorly understood. Heme acts as a physiological oxidant that triggers intense epimastigote proliferation and upregulates the expression of genes related to glycolysis and aerobic fermentation in vitro. Here, heme-cultured epimastigotes increased D-glucose consumption. In fact, heme-cultured parasites secreted more succinate (the end product of the so-called succinic fermentation) followed by glucose intake. Increased succinate levels reduced the extracellular pH, leading to acidification of the supernatant. However, the acidification and proliferation stimulated by heme was impaired when glycolysis was inhibited. Otherwise, when glucose amount is enhanced in supernatant, heme-cultured parasites increased its growth whereas the glucose depletion caused a delay in proliferation. Heme supplementation increased epimastigote electron transport system-related O2 consumption rates, while glucose addition reduced both the electron transport system-related O2 consumption rates and spare respiratory capacity, indicating a Crabtree-like effect. These results show that glycolysis predominated in heme-cultured epimastigotes over oxidative phosphorylation for energy supply when glucose is present to sustain its high proliferation in vitro. Furthermore, it provided an insight into the parasite biology in the vector environment that supply glucose and the digestion of blood generates free heme that can lead to the growth of T. cruzi epimastigotes.
Collapse
Affiliation(s)
- Carolina Silva Dias Vieira
- Laboratório de Interação Tripanossomatídeos e Vetores—Departamento de Bioquímica, IBRAG–UERJ–Rio de Janeiro, Brazil
| | - Ramon Pinheiro Aguiar
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM) and National Center for Structural Biology and Bioimaging (CENABIO)–UFRJ–Rio de Janeiro, Brazil
| | - Natalia Pereira de Almeida Nogueira
- Laboratório de Interação Tripanossomatídeos e Vetores—Departamento de Bioquímica, IBRAG–UERJ–Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia—Entomologia Molecular (INCT-EM)–Brazil
| | | | - Marcia Cristina Paes
- Laboratório de Interação Tripanossomatídeos e Vetores—Departamento de Bioquímica, IBRAG–UERJ–Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia—Entomologia Molecular (INCT-EM)–Brazil
| |
Collapse
|
37
|
Campbell PC, de Graffenried CL. Morphogenesis in Trypanosoma cruzi epimastigotes proceeds via a highly asymmetric cell division. PLoS Negl Trop Dis 2023; 17:e0011731. [PMID: 37917723 PMCID: PMC10656021 DOI: 10.1371/journal.pntd.0011731] [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: 06/05/2023] [Revised: 11/17/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023] Open
Abstract
Trypanosoma cruzi is a protist parasite that is the causative agent of Chagas disease, a neglected tropical disease endemic to the Americas. T. cruzi cells are highly polarized and undergo morphological changes as they cycle within their insect and mammalian hosts. Work on related trypanosomatids has described cell division mechanisms in several life-cycle stages and identified a set of essential morphogenic proteins that serve as markers for key events during trypanosomatid division. Here, we use Cas9-based tagging of morphogenic genes, live-cell imaging, and expansion microscopy to study the cell division mechanism of the insect-resident epimastigote form of T. cruzi, which represents an understudied trypanosomatid morphotype. We find that T. cruzi epimastigote cell division is highly asymmetric, producing one daughter cell that is significantly smaller than the other. Daughter cell division rates differ by 4.9 h, which may be a consequence of this size disparity. Many of the morphogenic proteins identified in T. brucei have altered localization patterns in T. cruzi epimastigotes, which may reflect fundamental differences in the cell division mechanism of this life cycle stage, which widens and shortens the cell body to accommodate the duplicated organelles and cleavage furrow rather than elongating the cell body along the long axis of the cell, as is the case in life-cycle stages that have been studied in T. brucei. This work provides a foundation for further investigations of T. cruzi cell division and shows that subtle differences in trypanosomatid cell morphology can alter how these parasites divide.
Collapse
Affiliation(s)
- Paul C. Campbell
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Christopher L. de Graffenried
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
38
|
Pacini MF, Perdomini A, Bulfoni Balbi C, Dinatale B, Herrera FE, Perez AR, Marcipar I. The high identity of the Trypanosoma cruzi Group-I of trans-sialidases points them as promising vaccine immunogens. Proteins 2023; 91:1444-1460. [PMID: 37323089 DOI: 10.1002/prot.26537] [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: 12/06/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Trans-sialidase (TS) superfamily of proteins comprises eight subgroups, being the proteins of Group-I (TS-GI) promising immunogens in vaccine approaches against Trypanosoma cruzi. Strikingly, TS-GI antigenic variability among parasite lineages and their influence on vaccine development has not been previously analyzed. Here, a search in GenBank detects 49 TS-GI indexed sequences, whereas the main infecting human different parasite discrete typing units (DTU) are represented. In silico comparison among these sequences indicate that they share an identity above 92%. Moreover, the antigenic regions (T-cell and B-cell epitopes) are conserved in most sequences or present amino acid substitutions that scarcely may alter the antigenicity. Additionally, since the generic term TS is usually used to refer to different immunogens of this broad family, a further in silico analysis of the TS-GI-derived fragments tested in preclinical vaccines was done to determine the coverage and identity among them, showing that overall amino acid identity of vaccine immunogens is high, but the segment coverage varies widely. Accordingly, strong H-2K, H-2I, and B-cell epitopes are dissimilarly represented among vaccine TS-derived fragments depending on the extension of the TG-GI sequence used. Moreover, bioinformatic analysis detected a set of 150 T-cell strong epitopes among the DTU-indexed sequences that strongly bind human HLA-I supertypes. In all currently reported experimental vaccines based on TS-GI fragments, mapping these 150 epitopes showed that they are moderately represented. However, despite vaccine epitopes do not present all the substitutions observed in the DTUs, these regions of the proteins are equally recognized by the same HLAs. Interestingly, the predictions regarding global and South American population coverage estimated in these 150 epitopes are similar to the estimations in experimental vaccines when the complete sequence of TS-GI is used as an antigen. In silico prediction also shows that a number of these MHC-I restricted T-cell strong epitopes could be also cross-recognized by HLA-I supertypes and H-2Kb or H-2Kd backgrounds, indicating that these mice may be used to improve and facilitate the development of new TS-based vaccines and suggesting an immunogenic and protective potential in humans. Further molecular docking analyses were performed to strengthen these results. Taken together, different strategies that would cover more or eventually fully of these T-cell and also B-cell epitopes to reach a high level of coverage are considered.
Collapse
Affiliation(s)
- Maria Florencia Pacini
- Laboratorio de Estudios en Enfermedad de Chagas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario, Argentina
| | - Adrián Perdomini
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Camila Bulfoni Balbi
- Laboratorio de Estudios en Enfermedad de Chagas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario, Argentina
| | - Brenda Dinatale
- Laboratorio de Estudios en Enfermedad de Chagas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario, Argentina
| | - Fernando E Herrera
- Área de Modelado Molecular, Departamento de Física, Facultad de Bioquímica y Ciencias, Universidad Nacional del Litoral, (CONICET), Santa Fe, Argentina
| | - Ana Rosa Perez
- Laboratorio de Estudios en Enfermedad de Chagas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Iván Marcipar
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
39
|
Marinov GK, Chen X, Swaffer MP, Xiang T, Grossman AR, Greenleaf WJ. Genome-wide distribution of 5-hydroxymethyluracil and chromatin accessibility in the Breviolum minutum genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558303. [PMID: 37781619 PMCID: PMC10541103 DOI: 10.1101/2023.09.18.558303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
In dinoflagellates, a unique and extremely divergent genomic and nuclear organization has evolved. The highly unusual features of dinoflagellate nuclei and genomes include permanently condensed liquid crystalline chromosomes, primarily packaged by proteins other than histones, genes organized in very long unidirectional gene arrays, a general absence of transcriptional regulation, high abundance of the otherwise very rare DNA modification 5-hydroxymethyluracil (5-hmU), and many others. While most of these fascinating properties were originally identified in the 1970s and 1980s, they have not yet been investigated using modern genomic tools. In this work, we address some of the outstanding questions regarding dinoflagellate genome organization by mapping the genome-wide distribution of 5-hmU (using both immunoprecipitation-based and basepair-resolution chemical mapping approaches) and of chromatin accessibility in the genome of the Symbiodiniaceae dinoflagellate Breviolum minutum. We find that the 5-hmU modification is preferentially enriched over certain classes of repetitive elements, often coincides with the boundaries between gene arrays, and is generally correlated with decreased chromatin accessibility, the latter otherwise being largely uniform along the genome. We discuss the potential roles of 5-hmU in the functional organization of dinoflagellate genomes and its relationship to the transcriptional landscape of gene arrays.
Collapse
Affiliation(s)
- Georgi K Marinov
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| | - Xinyi Chen
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA
| | | | - Tingting Xiang
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California 94305, USA
| | - Arthur R Grossman
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California 94305, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, California 94305, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, California 94305, USA
- Department of Applied Physics, Stanford University, Stanford, California 94305, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
40
|
Ur Rahman M, Khan M, Khan SW, Khan RU, Sohail A, Zaman A, Alam N. Novel Schiff bases of Vanillin: potent inhibitors of macrophage harbored Leishmania tropica. J Parasit Dis 2023; 47:619-629. [PMID: 37520206 PMCID: PMC10382424 DOI: 10.1007/s12639-023-01594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/05/2023] [Indexed: 08/01/2023] Open
Abstract
Due to limited chemotherapeutic options for leishmaniasis, novel synthetic compounds are gaining attention for evaluation against leishmaniasis. This study aimed to synthesize the compound's Schiff bases of Vanillin to investigate and evaluate their anti-leishmanial potentials against intracellular protozoan parasites Leishmania tropica. In the current study, the phenomena of synergism by designing Schiff bases with Vanillin enhances their desired importance. A total of five compounds Schiff bases of Vanillin were synthesized using different aromatic amines and Vanillin. The structural analysis of all the compounds was done through FT-IR (Fourier Transformer-Infrared), thin layer chromatography, and spectroscopic techniques such as 13C-NMR, mass spectrometry, and 1H-NMR. The antimicrobial properties of all the compounds ZI-1, ZI-2, BS-1, KH-1, and FA-2 against promastigotes and amastigotes forms of L. tropica were analyzed at three different concentrations 25, 50, and 100 µg/ml. The in-vitro MTT assay was performed to calculate the percent inhibition, IC50 values, and their cytotoxicity. The highest percent inhibition values against promastigote form of L. tropica were BS-1 53.78% at 25 µg/ml, ZI-2 66.95% at 50 µg/ml, and again ZI-2 76.92% at 100 µg/ml. Similarly, the highest percent inhibition values against intracellular amastigote stage were BS-1 55.77% at 25 µg/ml, ZI-2 67.78% at 50 µg/ml and again ZI-2 84.93% 100 µg/ml. The highest potency was recorded for BS-1 in both stages, with IC50 values of 9.83 and 4.27 µg/ml against promastigotes and intracellular amastigotes, respectively. The percent hemolysis as toxicity; the lowest percent hemolysis was recorded for ZI-1 at three different concentrations of 25, 50, 100 µg/ml of 2.60, 3.50, and 6.31, respectively. These results suggested that all the compounds exhibited anti-leishmanial activity, with BS-1 as the most potent. Further studies are suggested to increase the activity of compounds with structural modifications by the addition of some other synergistic, novel, and analogue compounds.
Collapse
Affiliation(s)
- Mujeeb Ur Rahman
- Department of Medical Microbiology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Momin Khan
- Department of Medical Microbiology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Sher Wali Khan
- Department of Chemistry, Shaheed Benazir Bhutto University, Sheringal, Khyber Pakhtunkhwa Pakistan
| | - Rahat Ullah Khan
- Department of Medical Microbiology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa Pakistan
- Faculty of Veterinary and Animal Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Aamir Sohail
- Department of Medical Microbiology, Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Ali Zaman
- Faculty of Veterinary and Animal Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Naveed Alam
- Faculty of Veterinary and Animal Sciences, Gomal University, Dera Ismail Khan, Pakistan
| |
Collapse
|
41
|
Chiurillo MA, Carlson J, Bertolini MS, Raja A, Lander N. Dual localization of receptor-type adenylate cyclases and cAMP response protein 3 unveils the presence of two putative signaling microdomains in Trypanosoma cruzi. mBio 2023; 14:e0106423. [PMID: 37477489 PMCID: PMC10470820 DOI: 10.1128/mbio.01064-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/02/2023] [Indexed: 07/22/2023] Open
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas disease, a leading cause of disability and premature death in the Americas. This parasite spends its life between a triatomine insect and a mammalian host, transitioning between developmental stages in response to microenvironmental changes. Among the second messengers driving differentiation in T. cruzi, cAMP has been shown to mediate metacyclogenesis and response to osmotic stress, but this signaling pathway remains largely unexplored in this parasite. Adenylate cyclases (ACs) catalyze the conversion of ATP to cAMP. They comprise a multigene family encoding putative receptor-type ACs in T. cruzi. Using protein sequence alignment, we classified them into five groups and chose a representative member from each group to study their localization (TcAC1-TcAC5). We expressed an HA-tagged version of each protein in T. cruzi and performed immunofluorescence analysis. A peculiar dual localization of TcAC1 and TcAC2 was observed in the flagellar distal domain and in the contractile vacuole complex (CVC), and their enzymatic activity was confirmed by gene complementation in yeast. Furthermore, TcAC1 overexpressing parasites showed an increased metacyclogenesis, a defect in host cell invasion, and a reduced intracellular replication, highlighting the importance of this protein throughout T. cruzi life cycle. These mutants were more tolerant to hypoosmotic stress and showed a higher adhesion capacity during in vitro metacyclogenesis, whereas the wild-type phenotype was restored after disrupting TcAC1 localization. Finally, TcAC1 was found to interact with cAMP response protein 3 (TcCARP3), co-localizing with this protein in the flagellar tip and CVC. IMPORTANCE We identified three components of the cAMP signaling pathway (TcAC1, TcAC2, and TcCARP3) with dual localization in Trypanosoma cruzi: the flagellar distal domain and the CVC, structures involved in cell adhesion and osmoregulation, respectively. We found evidence on the role of TcAC1 in both cellular processes, as well as in metacyclogenesis. Our data suggest that TcACs act as signal sensors and transducers through cAMP synthesis in membrane microdomains. We propose a model in which TcACs sense the harsh conditions in the triatomine hindgut (nutrient deprivation, acidic pH, osmotic stress, ionic composition, hydrophobic interactions) and become active. Synthesis of cAMP then triggers cell adhesion prior completion of metacyclogenesis, while mediating a response to osmotic stress in the parasite. These results shed light into the mechanisms driving cAMP-mediated cell differentiation in T. cruzi, while raising new questions on the activation of TcACs and the role of downstream components of this pathway.
Collapse
Affiliation(s)
- Miguel A. Chiurillo
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Joshua Carlson
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mayara S. Bertolini
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Aqsa Raja
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Noelia Lander
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Glockzin K, Meneely KM, Hughes R, Maatouk SW, Piña GE, Suthagar K, Clinch K, Buckler JN, Lamb AL, Tyler PC, Meek TD, Katzfuss A. Kinetic and Structural Characterization of Trypanosoma cruzi Hypoxanthine-Guanine-Xanthine Phosphoribosyltransferases and Repurposing of Transition-State Analogue Inhibitors. Biochemistry 2023. [PMID: 37418678 DOI: 10.1021/acs.biochem.3c00116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
Over 70 million people are currently at risk of developing Chagas Disease (CD) infection, with more than 8 million people already infected worldwide. Current treatments are limited and innovative therapies are required. Trypanosoma cruzi, the etiological agent of CD, is a purine auxotroph that relies on phosphoribosyltransferases to salvage purine bases from their hosts for the formation of purine nucleoside monophosphates. Hypoxanthine-guanine-xanthine phosphoribosyltransferases (HGXPRTs) catalyze the salvage of 6-oxopurines and are promising targets for the treatment of CD. HGXPRTs catalyze the formation of inosine, guanosine, and xanthosine monophosphates from 5-phospho-d-ribose 1-pyrophosphate and the nucleobases hypoxanthine, guanine, and xanthine, respectively. T. cruzi possesses four HG(X)PRT isoforms. We previously reported the kinetic characterization and inhibition of two isoforms, TcHGPRTs, demonstrating their catalytic equivalence. Here, we characterize the two remaining isoforms, revealing nearly identical HGXPRT activities in vitro and identifying for the first time T. cruzi enzymes with XPRT activity, clarifying their previous annotation. TcHGXPRT follows an ordered kinetic mechanism with a postchemistry event as the rate-limiting step(s) of catalysis. Its crystallographic structures reveal implications for catalysis and substrate specificity. A set of transition-state analogue inhibitors (TSAIs) initially developed to target the malarial orthologue were re-evaluated, with the most potent compound binding to TcHGXPRT with nanomolar affinity, validating the repurposing of TSAIs to expedite the discovery of lead compounds against orthologous enzymes. We identified mechanistic and structural features that can be exploited in the optimization of inhibitors effective against TcHGPRT and TcHGXPRT concomitantly, which is an important feature when targeting essential enzymes with overlapping activities.
Collapse
Affiliation(s)
- Kayla Glockzin
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| | - Kathleen M Meneely
- Department of Chemistry, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - Ryan Hughes
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| | - Sean W Maatouk
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| | - Grace E Piña
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| | - Kajitha Suthagar
- Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Keith Clinch
- Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Joshua N Buckler
- Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Audrey L Lamb
- Department of Chemistry, University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - Peter C Tyler
- Ferrier Research Institute, Victoria University of Wellington, Gracefield Research Centre, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Thomas D Meek
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| | - Ardala Katzfuss
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, Texas 77843-2128, United States
| |
Collapse
|
43
|
Gabaldón-Figueira JC, Martinez-Peinado N, Escabia E, Ros-Lucas A, Chatelain E, Scandale I, Gascon J, Pinazo MJ, Alonso-Padilla J. State-of-the-Art in the Drug Discovery Pathway for Chagas Disease: A Framework for Drug Development and Target Validation. Res Rep Trop Med 2023; 14:1-19. [PMID: 37337597 PMCID: PMC10277022 DOI: 10.2147/rrtm.s415273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
Chagas disease is the most important protozoan infection in the Americas, and constitutes a significant public health concern throughout the world. Development of new medications against its etiologic agent, Trypanosoma cruzi, has been traditionally slow and difficult, lagging in comparison with diseases caused by other kinetoplastid parasites. Among the factors that explain this are the incompletely understood mechanisms of pathogenesis of T. cruzi infection and its complex set of interactions with the host in the chronic stage of the disease. These demand the performance of a variety of in vitro and in vivo assays as part of any drug development effort. In this review, we discuss recent breakthroughs in the understanding of the parasite's life cycle and their implications in the search for new chemotherapeutics. For this, we present a framework to guide drug discovery efforts against Chagas disease, considering state-of-the-art preclinical models and recently developed tools for the identification and validation of molecular targets.
Collapse
Affiliation(s)
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Elisa Escabia
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - María-Jesús Pinazo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| |
Collapse
|
44
|
Ferpozzi H. Public-Private Partnerships and the Landscape of Neglected Tropical Disease Research: The Shifting Logic and Spaces of Knowledge Production. MINERVA 2023:1-23. [PMID: 37359299 PMCID: PMC10234793 DOI: 10.1007/s11024-023-09496-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/28/2023]
Abstract
Until the recent spread of public-private partnerships, pharmaceutical firms had avoided research and development into neglected tropical diseases (NTDs). Because these are diseases that affect the poorest populations in developing regions, research and development initiatives have for the most part depended on the resources and expertise drawn from academia, international organizations, and intermittent state interventions in disease-endemic countries. Over the last few decades, however, public-private product development partnerships (PDPs) have been introducing new collaborative agreements in which the existing resources and expertise combine with the those traditionally withheld by the pharmaceutical industry and global health NGOs. This paper explores recent transformations in the representation of NTDs by examining the shifting logic and spaces of knowledge production which the advent of PDPs has enabled. An analysis of two case studies focused on Chagas disease-related initiatives addresses recurring preoccupations in Science, Technology and Society studies as well as in critical analyses of PDPs: that is, the back-and-forth movement of the disease from being an object of scientific inquiry to a public health concern, and the legitimacy risks and material asymmetries entailed in global health PDPs. Both cases show that it is major global health stakeholders and experts in non-endemic countries, rather than transnational pharmaceutical firms, that exert the greatest influence upon these changing representations: PDPs attempt to expand the preexisting biomedical focus on NTDs by means of incorporating "real world" drug development preoccupations (which I term epistemic shifts), but they also combine their stated global humanitarian aim with security concerns about the diseases spreading to non-endemic, industrialized countries (which I term geographical shifts).
Collapse
Affiliation(s)
- Hugo Ferpozzi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Centro de Ciencia, Tecnología y Sociedad, Universidad Maimónides, Buenos Aires, Argentina
| |
Collapse
|
45
|
Verçosa BLA, Muniz-Junqueira MI, Barradas ALB, Costa FAL, Melo MN, Vasconcelos AC. Enhanced apoptotic index in hepatocytes, Kupffer cells, and inflammatory infiltrate showed positive correlation with hepatic lesion intensity, parasite load, and clinical status in naturally Leishmania-infected dogs. Microb Pathog 2023:106194. [PMID: 37269879 DOI: 10.1016/j.micpath.2023.106194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
It is unknown if Leishmania amastigote infections affect hepatocytes and Kupffer cell apoptosis, and the role played by apoptosis in liver lesions in leishmaniasis is still unclear. Clinically affected and subclinically infected dogs with leishmaniosis and uninfected controls were assessed. Parasite load, biochemical markers for evaluation of liver damage, morphometry (area, perimeter, number of inflammatory focus, major and minor diameters), apoptosis in hepatic tissue (hepatocytes, Kupffer cells, and inflammatory infiltrates) and cellularity in inflammatory foci were quantified. The parasite load in clinically affected dogs proved to be higher than in the other groups. All morphometric parameters (area, perimeter, number of inflammatory focus, major and minor diameters) from clinically affected were higher than the values found in the subclinically infected and uninfected control dogs. Only clinically affected dogs presented high levels of ALT, FA, GGT and cholesterol in serum. Strong positive correlation was observed between biochemical markers for evaluation of liver damage (ALT, FA, GGT and cholesterol) and hepatic apoptosis (hepatocytes, Kupffer cells, and inflammation). Clinically affected dogs showed a more intense hepatic lesion. Hepatocytes showed a higher rate of apoptosis in Leishmania-infected dogs than in uninfected control dogs. The Kupffer cell apoptotic index and apoptosis within the inflammatory infiltrates were higher in clinically affected dogs. The apoptotic index evaluated in hepatocytes, Kupffer cells, and inflammatory infiltrates showed a positive correlation with the intensity of the hepatic lesion, parasite load, and clinical status. Apoptotic cells also showed positive immunostaining for TUNEL, Bcl2, and Bax. Our data showed that hepatic apoptosis was related to the severity of liver damage, the progression of infection, and the parasite load in leishmaniasis. Apoptotic regulated cell recruitment modulated the inflammatory response and favored the survival and dissemination of parasites, depending on the clinical status of the Leishmania-infected dogs.
Collapse
Affiliation(s)
- Bárbara Laurice Araujo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil; Faculdade de Ciências da Saúde Pitágoras de Codó, Maranhão, Brazil.
| | | | - Ana Lys Bezerra Barradas
- Departamento de Clínica e Cirurgia veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Francisco Assis Lima Costa
- Departamento de Clínica e Cirurgia veterinária, Centro de Ciências Agrárias, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
46
|
Campbell PC, de Graffenried CL. Morphogenesis in Trypanosoma cruzi epimastigotes proceeds via a highly asymmetric cell division. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542100. [PMID: 37293088 PMCID: PMC10245916 DOI: 10.1101/2023.05.24.542100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Trypanosoma cruzi is a protist parasite that is the causative agent of Chagas' disease, a neglected tropical disease endemic to the Americas. T. cruzi cells are highly polarized and undergo morphological changes as they cycle within their insect and mammalian hosts. Work on related trypanosomatids has described cell division mechanisms in several life-cycle stages and identified a set of essential morphogenic proteins that serve as markers for key events during trypanosomatid division. Here, we use Cas9-based tagging of morphogenic genes, live-cell imaging, and expansion microscopy to study the cell division mechanism of the insect-resident epimastigote form of T. cruzi, which represents an understudied trypanosomatid morphotype. We find that T. cruzi epimastigote cell division is highly asymmetric, producing one daughter cell that is significantly smaller than the other. Daughter cell division rates differ by 4.9 h, which may be a consequence of this size disparity. Many of the morphogenic proteins identified in T. brucei have altered localization patterns in T. cruzi epimastigoes, which may reflect fundamental differences in the cell division mechanism of this life cycle stage, which widens and shortens the cell body to accommodate the duplicated organelles and cleavage furrow rather than elongating the cell body along the long axis of the cell, as is the case in life-cycle stages that have been studied in T. brucei. This work provides a foundation for further investigations of T. cruzi cell division and shows that subtle differences in trypansomatid cell morphology can alter how these parasites divide.
Collapse
Affiliation(s)
- Paul C. Campbell
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
47
|
Soprano LL, Ferrero MR, Jacobs T, Couto AS, Duschak VG. Hallmarks of the relationship between host and Trypanosoma cruzi sulfated glycoconjugates along the course of Chagas disease. Front Cell Infect Microbiol 2023; 13:1028496. [PMID: 37256110 PMCID: PMC10225527 DOI: 10.3389/fcimb.2023.1028496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
American Trypanosomiasis or Chagas disease (ChD), a major problem that is still endemic in large areas of Latin America, is caused by Trypanosoma cruzi. This agent holds a major antigen, cruzipain (Cz). Its C-terminal domain (C-T) is retained in the glycoprotein mature form and bears several post-translational modifications. Glycoproteins containing sulfated N-linked oligosaccharides have been mostly implicated in numerous specific procedures of molecular recognition. The presence of sulfated oligosaccharides was demonstrated in Cz, also in a minor abundant antigen with serine-carboxypeptidase (SCP) activity, as well as in parasite sulfatides. Sulfate-bearing glycoproteins in Trypanosomatids are targets of specific immune responses. T. cruzi chronically infected subjects mount specific humoral immune responses to sulfated Cz. Unexpectedly, in the absence of infection, mice immunized with C-T, but not with sulfate-depleted C-T, showed ultrastructural heart anomalous pathological effects. Moreover, the synthetic anionic sugar conjugate GlcNAc6SO3-BSA showed to mimic the N-glycan-linked sulfated epitope (sulfotope) humoral responses that natural Cz elicits. Furthermore, it has been reported that sulfotopes participate via the binding of sialic acid Ig-like-specific lectins (Siglecs) to sulfosialylated glycoproteins in the immunomodulation by host-parasite interaction as well as in the parasite infection process. Strikingly, recent evidence involved Cz-sulfotope-specific antibodies in the immunopathogenesis and infection processes during the experimental ChD. Remarkably, sera from chronically T. cruzi-infected individuals with mild disease displayed higher levels of IgG2 antibodies specific for sulfated glycoproteins and sulfatides than those with more severe forms of the disease, evidencing that T. cruzi sulfotopes are antigenic independently of the sulfated glycoconjugate type. Ongoing assays indicate that antibodies specific for sulfotopes might be considered biomarkers of human cardiac ChD progression, playing a role as predictors of stability from the early mild stages of chronic ChD.
Collapse
Affiliation(s)
- Luciana L. Soprano
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Maximiliano R. Ferrero
- Max-Planck Heart and Lung Laboratory, Research Institute in Biomedicine in Buenos Aires (IBioBA), Argentine-Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
| | - Thomas Jacobs
- Immunology Department, Bernhard Notch Institute of Tropical Medicine, Hamburg, Germany
| | - Alicia S. Couto
- Faculty in Exact and Natural Sciences (FCEN), Chemical Organic Department-National Council of Scientific and Technical Research (CONICET), Center of CarboHydrates (CHIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina
| | - Vilma G. Duschak
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
48
|
Ricci AD, Bracco L, Salas-Sarduy E, Ramsey JM, Nolan MS, Lynn MK, Altcheh J, Ballering GE, Torrico F, Kesper N, Villar JC, Marcipar IS, Marco JD, Agüero F. The Trypanosoma cruzi Antigen and Epitope Atlas: antibody specificities in Chagas disease patients across the Americas. Nat Commun 2023; 14:1850. [PMID: 37012236 PMCID: PMC10070320 DOI: 10.1038/s41467-023-37522-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
During an infection the immune system produces pathogen-specific antibodies. These antibody repertoires become specific to the history of infections and represent a rich source of diagnostic markers. However, the specificities of these antibodies are mostly unknown. Here, using high-density peptide arrays we examined the human antibody repertoires of Chagas disease patients. Chagas disease is a neglected disease caused by Trypanosoma cruzi, a protozoan parasite that evades immune mediated elimination and mounts long-lasting chronic infections. We describe a proteome-wide search for antigens, characterised their linear epitopes, and show their reactivity on 71 individuals from diverse human populations. Using single-residue mutagenesis we revealed the core functional residues for 232 of these epitopes. Finally, we show the diagnostic performance of identified antigens on challenging samples. These datasets enable the study of the Chagas antibody repertoire at an unprecedented depth and granularity, while also providing a rich source of serological biomarkers.
Collapse
Affiliation(s)
- Alejandro D Ricci
- Instituto de Investigaciones Biotecnológicas (IIB) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Leonel Bracco
- Instituto de Investigaciones Biotecnológicas (IIB) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Emir Salas-Sarduy
- Instituto de Investigaciones Biotecnológicas (IIB) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina
| | - Janine M Ramsey
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública, Tapachula, México
| | - Melissa S Nolan
- Laboratory of Vector-borne and Zoonotic Diseases, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - M Katie Lynn
- Laboratory of Vector-borne and Zoonotic Diseases, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jaime Altcheh
- Hospital de Niños "Ricardo Gutierrez", Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP) - GCBA-CONICET, Buenos Aires, Argentina
| | - Griselda E Ballering
- Hospital de Niños "Ricardo Gutierrez", Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | | | - Norival Kesper
- LIM-49, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Juan C Villar
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga y Fundación Cardioinfantil - Instituto de Cardiología, Bogotá, Colombia
| | - Iván S Marcipar
- Facultad de Ciencias Médicas y Facultad de Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Jorge D Marco
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina.
- Escuela de Bio y Nanotecnologías (EByN), Universidad de San Martín (UNSAM), San Martín, Buenos Aires, Argentina.
| |
Collapse
|
49
|
Behind Base J: The Roles of JBP1 and JBP2 on Trypanosomatids. Pathogens 2023; 12:pathogens12030467. [PMID: 36986389 PMCID: PMC10057400 DOI: 10.3390/pathogens12030467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
β-D-glucopyranosyloxymethiluracil (Base J) is a modified thymidine base found in kinetoplastids and some related organisms. Interestingly, Base J distribution into the genome can vary depending on the organism and its life stage. Base J is reported to be found mostly at telomeric repeats, on inactive variant surface glycoproteins (VSG’s) expression sites (e.g., T. brucei), in RNA polymerase II termination sites and sub-telomeric regions (e.g., Leishmania). This hypermodified nucleotide is synthesized in two steps with the participation of two distinct thymidine hydroxylases, J-binding protein 1 and 2 (JBP1 and JBP2, respectively) and a β-glucosyl transferase. A third J-binding protein, named JBP3, was recently identified as part of a multimeric complex. Although its structural similarities with JBP1, it seems not to be involved in J biosynthesis but to play roles in gene expression regulation in trypanosomatids. Over the years, with the characterization of JBP1 and JBP2 mutant lines, Base J functions have been targeted and shone a light on that matter, showing genus-specific features. This review aims to explore Base J’s reported participation as a regulator of RNA polymerase II transcription termination and to summarize the functional and structural characteristics and similarities of the remarkable JBP proteins in pathogenic trypanosomatids.
Collapse
|
50
|
Urán Landaburu L, Didier Garnham M, Agüero F. Targeting trypanosomes: how chemogenomics and artificial intelligence can guide drug discovery. Biochem Soc Trans 2023; 51:195-206. [PMID: 36606702 DOI: 10.1042/bst20220618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
Trypanosomatids are protozoan parasites that cause human and animal neglected diseases. Despite global efforts, effective treatments are still much needed. Phenotypic screens have provided several chemical leads for drug discovery, but the mechanism of action for many of these chemicals is currently unknown. Recently, chemogenomic screens assessing the susceptibility or resistance of parasites carrying genome-wide modifications started to define the mechanism of action of drugs at large scale. In this review, we discuss how genomics is being used for drug discovery in trypanosomatids, how integration of chemical and genomics data from these and other organisms has guided prioritisations of candidate therapeutic targets and additional chemical starting points, and how these data can fuel the expansion of drug discovery pipelines into the era of artificial intelligence.
Collapse
Affiliation(s)
- Lionel Urán Landaburu
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Mercedes Didier Garnham
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
- Escuela de Bio y Nanociencias (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| |
Collapse
|