1
|
Ernst L, Macedo GC, McCall LI. System-based insights into parasitological and clinical treatment failure in Chagas disease. mSystems 2025; 10:e0003824. [PMID: 39772644 PMCID: PMC11834445 DOI: 10.1128/msystems.00038-24] [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] [Indexed: 01/11/2025] Open
Abstract
Infectious disease treatment success requires symptom resolution (clinical treatment success), which often but not always involves pathogen clearance. Both of these treatment goals face disease-specific and general challenges. In this review, we summarize the current state of knowledge in mechanisms of clinical and parasitological treatment failure in the context of Chagas disease, a neglected tropical disease causing cardiac and gastrointestinal symptoms. Parasite drug resistance and persistence, drug pharmacokinetics and dynamics, as well as persistently altered host immune responses and tissue damage are the most common reasons for Chagas disease treatment failure. We discuss the therapeutics that failed before regulatory approval, limitations of current therapeutic options and new treatment strategies to overcome persistent parasites, inflammatory responses, and metabolic alterations. Large-scale omics analyses were critical in generating these insights and will continue to play a prominent role in addressing the challenges still facing Chagas disease drug treatment.
Collapse
Affiliation(s)
- Luis Ernst
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| | - Giovana C. Macedo
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
2
|
Aldfer MM, Hulpia F, van Calenbergh S, De Koning HP. Mapping the transporter-substrate interactions of the Trypanosoma cruzi NB1 nucleobase transporter reveals the basis for its high affinity and selectivity for hypoxanthine and guanine and lack of nucleoside uptake. Mol Biochem Parasitol 2024; 258:111616. [PMID: 38401850 DOI: 10.1016/j.molbiopara.2024.111616] [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: 11/20/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Trypanosoma cruzi is a protozoan parasite and the etiological agent of Chagas disease, a debilitating and sometimes fatal disease that continues to spread to new areas. Yet, Chagas disease is still only treated with two related nitro compounds that are insufficiently effective and cause severe side effects. Nucleotide metabolism is one of the known vulnerabilities of T. cruzi, as they are auxotrophic for purines, and nucleoside analogues have been shown to have genuine promise against this parasite in vitro and in vivo. Since purine antimetabolites require efficient uptake through transporters, we here report a detailed characterisation of the T. cruzi NB1 nucleobase transporter with the aim of elucidating the interactions between TcrNB1 and its substrates and finding the positions that can be altered in the design of novel antimetabolites without losing transportability. Systematically determining the inhibition constants (Ki) of purine analogues for TcrNB1 yielded their Gibbs free energy of interaction, ΔG0. Pairwise comparisons of substrate (hypoxanthine, guanine, adenine) and analogues allowed us to determine that optimal binding affinity by TcrNB1 requires interactions with all four nitrogen residues of the purine ring, with N1 and N9, in protonation state, functioning as presumed hydrogen bond donors and unprotonated N3 and N7 as hydrogen bond acceptors. This is the same interaction pattern as we previously described for the main nucleobase transporters of Trypanosoma brucei spp. and Leishmania major and makes it the first of the ENT-family genes that is functionally as well as genetically conserved between the three main kinetoplast pathogens.
Collapse
Affiliation(s)
- Mustafa M Aldfer
- School of Infection and Immunity, Sir Graeme Davies Building, 120 University Place, University of Glasgow, GlasgowG12 8TA, UK
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, 9000 Gent, Belgium
| | - Serge van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, 9000 Gent, Belgium
| | - Harry P De Koning
- School of Infection and Immunity, Sir Graeme Davies Building, 120 University Place, University of Glasgow, GlasgowG12 8TA, UK.
| |
Collapse
|
3
|
Olanlokun JO, Abiodun OW, Adegbuyi AT, Koorbanally NA, Olorunsogo OO. Mefloquine-curcumin combinations improve host mitochondrial respiration and decrease mitotoxic effects of mefloquine in Plasmodium berghei-infected mice. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 6:100180. [PMID: 38725654 PMCID: PMC11081784 DOI: 10.1016/j.crphar.2024.100180] [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: 11/22/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 05/12/2024] Open
Abstract
Plasmodium infection is a health challenge. Although, antiplasmodial drugs kill the parasites, information on the effects of infection and drugs on the expression of some genes is limited. Malaria was induced in two different studies using NK65 (chloroquine-susceptible, study 1), and ANKA (chloroquine-resistant, study 2) strains of Plasmodium berghei in 30 male Swiss mice (n = 5) in each study. Mice orally received 10 mL/kg distilled water, (infected control), Mefloquine (MF) (10 mg/kg), MF and Curcumin (CM) (25 mg/kg), MF and CM (50 mg/kg), CM (25 mg/kg) and CM (50 mg/kg). Five mice (un-infected) were used as the control. After treatment, total Ribonucleic acid (RNA) was isolated from liver and erythrocytes while Deoxyribonucleic acid (DNA)-free RNA were converted to cDNA. Polymerase Chain Reaction (PCR) amplification was performed and relative expressions of FIKK12, AQP3, P38 MAPK, NADH oxidoreductase, and cytochrome oxidase expressions were determined. Markers of glycolysis, toxicity and antioxidants were determined using ELISA assays. While the expression of FIKK12 was blunted by MF in the susceptible study, co-treatment with curcumin (25 mg/kg) yielded the same results in the chloroquine-resistant study. Similar results were obtained on AQP3 in both studies. Curcumin decreased P38 MAPK in both studies. Plasmodium infection decreased NADH oxidoreductase and cytochrome oxidase but mefloquine-curcumin restored the expression of these genes. While glycolysis and toxicity were inhibited, antioxidant systems improved in the treated groups. Curcumin is needed for effective therapeutic efficacy and prevention of toxicity. Plasmodium infection and treatment modulate the expressions of some genes in the host. Curcumin combination with mefloquine modulates the expression of some genes in the host.
Collapse
Affiliation(s)
- John Oludele Olanlokun
- Laboratories for Biomemebrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oshireku Wisdom Abiodun
- Laboratories for Biomemebrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Neil Anthony Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000, South Africa
| | - Olufunso Olabode Olorunsogo
- Laboratories for Biomemebrane Research and Biotechnology, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
4
|
Carey SM, O’Neill DM, Conner GB, Sherman J, Rodriguez A, D’Antonio EL. Discovery of Strong 3-Nitro-2-Phenyl- 2H-Chromene Analogues as Antitrypanosomal Agents and Inhibitors of Trypanosoma cruzi Glucokinase. Int J Mol Sci 2024; 25:4319. [PMID: 38673904 PMCID: PMC11050443 DOI: 10.3390/ijms25084319] [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: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.
Collapse
Affiliation(s)
- Shane M. Carey
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Destiny M. O’Neill
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Garrett B. Conner
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| |
Collapse
|
5
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
6
|
Kwakye-Nuako G, Middleton CE, McCall LI. Small molecule mediators of host-T. cruzi-environment interactions in Chagas disease. PLoS Pathog 2024; 20:e1012012. [PMID: 38457443 PMCID: PMC10923493 DOI: 10.1371/journal.ppat.1012012] [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] [Indexed: 03/10/2024] Open
Abstract
Small molecules (less than 1,500 Da) include major biological signals that mediate host-pathogen-microbiome communication. They also include key intermediates of metabolism and critical cellular building blocks. Pathogens present with unique nutritional needs that restrict pathogen colonization or promote tissue damage. In parallel, parts of host metabolism are responsive to immune signaling and regulated by immune cascades. These interactions can trigger both adaptive and maladaptive metabolic changes in the host, with microbiome-derived signals also contributing to disease progression. In turn, targeting pathogen metabolic needs or maladaptive host metabolic changes is an important strategy to develop new treatments for infectious diseases. Trypanosoma cruzi is a single-celled eukaryotic pathogen and the causative agent of Chagas disease, a neglected tropical disease associated with cardiac and intestinal dysfunction. Here, we discuss the role of small molecules during T. cruzi infection in its vector and in the mammalian host. We integrate these findings to build a theoretical interpretation of how maladaptive metabolic changes drive Chagas disease and extrapolate on how these findings can guide drug development.
Collapse
Affiliation(s)
- Godwin Kwakye-Nuako
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Caitlyn E. Middleton
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| |
Collapse
|
7
|
Morrison LJ, Steketee PC, Tettey MD, Matthews KR. Pathogenicity and virulence of African trypanosomes: From laboratory models to clinically relevant hosts. Virulence 2023; 14:2150445. [PMID: 36419235 DOI: 10.1080/21505594.2022.2150445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
African trypanosomes are vector-borne protozoa, which cause significant human and animal disease across sub-Saharan Africa, and animal disease across Asia and South America. In humans, infection is caused by variants of Trypanosoma brucei, and is characterized by varying rate of progression to neurological disease, caused by parasites exiting the vasculature and entering the brain. Animal disease is caused by multiple species of trypanosome, primarily T. congolense, T. vivax, and T. brucei. These trypanosomes also infect multiple species of mammalian host, and this complexity of trypanosome and host diversity is reflected in the spectrum of severity of disease in animal trypanosomiasis, ranging from hyperacute infections associated with mortality to long-term chronic infections, and is also a main reason why designing interventions for animal trypanosomiasis is so challenging. In this review, we will provide an overview of the current understanding of trypanosome determinants of infection progression and severity, covering laboratory models of disease, as well as human and livestock disease. We will also highlight gaps in knowledge and capabilities, which represent opportunities to both further our fundamental understanding of how trypanosomes cause disease, as well as facilitating the development of the novel interventions that are so badly needed to reduce the burden of disease caused by these important pathogens.
Collapse
Affiliation(s)
- Liam J Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Pieter C Steketee
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Mabel D Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
8
|
Tan X, Yan Y, Zhang G, Li P, Ling F, Liu T, Wang G. Target epidermal damage of Gyrodactylus kobayashii to obtain the effective anthelmintic compound glacial acetic acid. AQUACULTURE 2023; 577:739993. [DOI: 10.1016/j.aquaculture.2023.739993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
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
|
10
|
Jiang K, Bai R, Gao T, Lu P, Zhang J, Zhang S, Xu F, Wang S, Zhao H. Optimization of hydrogen production in Enterobacter aerogenes by Complex I peripheral fragments destruction and maeA overexpression. Microb Cell Fact 2023; 22:137. [PMID: 37496040 PMCID: PMC10373349 DOI: 10.1186/s12934-023-02155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
As a concentrated energy source with high added value, hydrogen has great development prospects, with special emphasis on sustainable microbial production as a replacement for traditional fossil fuels. In this study, λ-Red recombination was used to alter the activity of Complex I by single and combined knockout of nuoE, nuoF and nuoG. In addition, the conversion of malic to pyruvic acid was promoted by overexpressing the maeA gene, which could increase the content of NADH and formic acid in the bacterial cells. Compared to the original strain, hydrogen production was 65% higher in the optimized strain IAM1183-EFG/M, in which the flux of the formic acid pathway was increased by 257%, the flux of the NADH pathway was increased by 13%, and the content of metabolites also changed significantly. In further bioreactor, the total hydrogen production of the scale-up IAM1183-EFG/M after 44 h of fermentation was 4.76 L, which increased by 18% compared with the starting strain. This study provides a new direction for future exploration of microbial hydrogen production by combinatorial modification of multiple genes.
Collapse
Affiliation(s)
- Ke Jiang
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ruoxuan Bai
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ting Gao
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ping Lu
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jingya Zhang
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Shuting Zhang
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Fangxu Xu
- Shenyang Functional Cordyceps militaris Industrial Technology Research Institute, Shenyang, 110034, China
- Liaoning Province Key Laboratory of Cordyceps Militaris with Functional Value, Experimental Teaching Center, Shenyang Normal University, Shenyang, 110034, China
| | - Shenghou Wang
- Shenyang Functional Cordyceps militaris Industrial Technology Research Institute, Shenyang, 110034, China
- Liaoning Province Key Laboratory of Cordyceps Militaris with Functional Value, Experimental Teaching Center, Shenyang Normal University, Shenyang, 110034, China
| | - Hongxin Zhao
- Zhejiang Province Key Laboratory of Plant Secondary Metabolism and Regulation, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
11
|
Nasuhidehnavi A, McCall LI. It takes two to tango: How immune responses and metabolic changes jointly shape cardiac Chagas disease. PLoS Pathog 2023; 19:e1011399. [PMID: 37262078 PMCID: PMC10234536 DOI: 10.1371/journal.ppat.1011399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Affiliation(s)
- Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, United States of America
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, United States of America
| |
Collapse
|
12
|
Won MM, Krüger T, Engstler M, Burleigh BA. The Intracellular Amastigote of Trypanosoma cruzi Maintains an Actively Beating Flagellum. mBio 2023; 14:e0355622. [PMID: 36840555 PMCID: PMC10128032 DOI: 10.1128/mbio.03556-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Throughout its complex life cycle, the uniflagellate parasitic protist, Trypanosoma cruzi, adapts to different host environments by transitioning between elongated motile extracellular stages and a nonmotile intracellular amastigote stage that replicates in the cytoplasm of mammalian host cells. Intracellular T. cruzi amastigotes retain a short flagellum that extends beyond the opening of the flagellar pocket with access to the extracellular milieu. Contrary to the long-held view that the T. cruzi amastigote flagellum is inert, we report that this organelle is motile and displays quasiperiodic beating inside mammalian host cells. Kymograph analysis determined an average flagellar beat frequency of ~0.7 Hz for intracellular amastigotes and similar beat frequencies for extracellular amastigotes following their isolation from host cells. Inhibitor studies reveal that flagellar motility in T. cruzi amastigotes is critically dependent on parasite mitochondrial oxidative phosphorylation. These novel observations reveal that flagellar motility is an intrinsic property of T. cruzi amastigotes and suggest that this organelle may play an active role in the parasite infection process. IMPORTANCE Understanding the interplay between intracellular pathogens and their hosts is vital to the development of new treatments and preventive strategies. The intracellular "amastigote" stage of the Chagas disease parasite, Trypanosoma cruzi, is a critical but understudied parasitic life stage. Previous work established that cytosolically localized T. cruzi amastigotes engage physically and selectively with host mitochondria using their short, single flagellum. The current study was initiated to examine the dynamics of the parasite flagellum-host mitochondrial interaction through live confocal imaging and led to the unexpected discovery that the T. cruzi amastigote flagellum is motile.
Collapse
Affiliation(s)
- Madalyn M. Won
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Timothy Krüger
- Department of Cell and Developmental Biology, Biozentrum, University of Würzburg, Germany
| | - Markus Engstler
- Department of Cell and Developmental Biology, Biozentrum, University of Würzburg, Germany
| | - Barbara A. Burleigh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Rojas-Pirela M, Kemmerling U, Quiñones W, Michels PAM, Rojas V. Antimicrobial Peptides (AMPs): Potential Therapeutic Strategy against Trypanosomiases? Biomolecules 2023; 13:biom13040599. [PMID: 37189347 DOI: 10.3390/biom13040599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Trypanosomiases are a group of tropical diseases that have devastating health and socio-economic effects worldwide. In humans, these diseases are caused by the pathogenic kinetoplastids Trypanosoma brucei, causing African trypanosomiasis or sleeping sickness, and Trypanosoma cruzi, causing American trypanosomiasis or Chagas disease. Currently, these diseases lack effective treatment. This is attributed to the high toxicity and limited trypanocidal activity of registered drugs, as well as resistance development and difficulties in their administration. All this has prompted the search for new compounds that can serve as the basis for the development of treatment of these diseases. Antimicrobial peptides (AMPs) are small peptides synthesized by both prokaryotes and (unicellular and multicellular) eukaryotes, where they fulfill functions related to competition strategy with other organisms and immune defense. These AMPs can bind and induce perturbation in cell membranes, leading to permeation of molecules, alteration of morphology, disruption of cellular homeostasis, and activation of cell death. These peptides have activity against various pathogenic microorganisms, including parasitic protists. Therefore, they are being considered for new therapeutic strategies to treat some parasitic diseases. In this review, we analyze AMPs as therapeutic alternatives for the treatment of trypanosomiases, emphasizing their possible application as possible candidates for the development of future natural anti-trypanosome drugs.
Collapse
|
14
|
Osorio-Méndez JF, Téllez GA, Zapata-López D, Echeverry S, Castaño JC. Sequence analysis of SWEET transporters from trypanosomatids and evaluation of its expression in Trypanosoma cruzi. Exp Parasitol 2023; 248:108496. [PMID: 36878387 DOI: 10.1016/j.exppara.2023.108496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/31/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Trypanosoma cruzi is an obligate parasite that uses glucose as one of the main resources to maintain its survival and proliferation. In eukaryotic cells glucose transport across membranes is mediated by facilitated transport through a variety of transporters. Herein, genes from the recently described SWEET family of carbohydrate transporters were identified in trypanosomatid parasites, including the medically important species T. cruzi and Leishmania spp. The identified genes have sequences with the typical attributes of known SWEET transporters. The expression of TcSWEET, the gene for the SWEET transporter found in the T. cruzi genome, was evidenced by immunohistochemistry using a polyclonal serum raised against peptides selected from the deduced TcSWEET protein sequence. In Western blot analysis, this α-TcSWEET serum detected proteins within the theoretical molecular mass for TcSWEET (25.8 kDa) in total epimastigote lysates, suggesting its expression at this parasite stage. Additionally, this serum stained epimastigotes at localizations consistent with the cell body and the flagellum. Together, these data suggests that SWEET transporters may contribute to glucose transport in trypanosomatid parasites.
Collapse
Affiliation(s)
- Juan Felipe Osorio-Méndez
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia.
| | - Germán Alberto Téllez
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Daniela Zapata-López
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Sebastián Echeverry
- Laboratorio de Microbiología y Biología Molecular, Programa de Medicina, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia, Colombia
| | - Jhon Carlos Castaño
- Grupo de Inmunología Molecular (Gymol), Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Quindío, Colombia
| |
Collapse
|
15
|
Ndlovu IS, Silas E, Tshilwane SI, Chaisi M, Vosloo A, Mukaratirwa S. Preliminary insights on the metabolomics of Trichinella zimbabwensis infection in Sprague Dawley rats using GCxGC-TOF-MS (untargeted approach). Front Mol Biosci 2023; 10:1128542. [PMID: 36876045 PMCID: PMC9983363 DOI: 10.3389/fmolb.2023.1128542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Trichinella infections have been documented globally and have been detected in wild and/or domestic animals except Antarctica. There is paucity of information in the metabolic responses of hosts during Trichinella infections and biomarkers for infection that can be used in the diagnosis of the disease. The current study aimed to apply a non-targeted metabolomic approach to identify Trichinella zimbabwensis biomarkers including metabolic response from sera of infected Sprague-Dawley rats. Fifty-four male Sprague-Dawley rats were randomly assigned into T. zimbabwensis infected group (n = 36) and the non-infected control (n = 18). Results from the study showed that the metabolic signature of T. zimbabwensis infection consists of enriched methyl histidine metabolism, disturbance of the liver urea cycle, impeded TCA cycle, and upregulation of gluconeogenesis metabolism. The observed disturbance in the metabolic pathways was attributed to the effects caused by the parasite during its migration to the muscles resulting in downregulation of amino acids intermediates in the Trichinella-infected animals, and therefore affecting energy production and degradation of biomolecules. It was concluded that T. zimbabwensis infection caused an upregulation of amino acids; pipecolic acid, histidine, and urea, and upregulation of glucose and meso-Erythritol. Moreover, T. zimbabwensis infection caused upregulation of the fatty acids, retinoic acid, and acetic acid. These findings highlight the potential of metabolomics as a novel approach for fundamental investigations of host-pathogen interactions as well as for disease progression and prognosis.
Collapse
Affiliation(s)
- I. S. Ndlovu
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Ekuyikeno Silas
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - S. I. Tshilwane
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - M. Chaisi
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
- Foundational Biodiversity Science, South African National Biodiversity Institute, Pretoria, South Africa
| | - A. Vosloo
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - S. Mukaratirwa
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
- One Health Center for Zoonoses and Tropical Veterinary Medicine, School of Veterinary Medicine, Ross University, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
16
|
Saraiva FMS, Cosentino-Gomes D, Inacio JDF, Almeida-Amaral EE, Louzada-Neto O, Rossini A, Nogueira NP, Meyer-Fernandes JR, Paes MC. Hypoxia Effects on Trypanosoma cruzi Epimastigotes Proliferation, Differentiation, and Energy Metabolism. Pathogens 2022; 11:pathogens11080897. [PMID: 36015018 PMCID: PMC9416468 DOI: 10.3390/pathogens11080897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022] Open
Abstract
Trypanosoma cruzi, the causative agent of Chagas disease, faces changes in redox status and nutritional availability during its life cycle. However, the influence of oxygen fluctuation upon the biology of T. cruzi is unclear. The present work investigated the response of T. cruzi epimastigotes to hypoxia. The parasites showed an adaptation to the hypoxic condition, presenting an increase in proliferation and a reduction in metacyclogenesis. Additionally, parasites cultured in hypoxia produced more reactive oxygen species (ROS) compared to parasites cultured in normoxia. The analyses of the mitochondrial physiology demonstrated that hypoxic condition induced a decrease in both oxidative phosphorylation and mitochondrial membrane potential (ΔΨm) in epimastigotes. In spite of that, ATP levels of parasites cultivated in hypoxia increased. The hypoxic condition also increased the expression of the hexokinase and NADH fumarate reductase genes and reduced NAD(P)H, suggesting that this increase in ATP levels of hypoxia-challenged parasites was a consequence of increased glycolysis and fermentation pathways. Taken together, our results suggest that decreased oxygen levels trigger a shift in the bioenergetic metabolism of T. cruzi epimastigotes, favoring ROS production and fermentation to sustain ATP production, allowing the parasite to survive and proliferate in the insect vector.
Collapse
Affiliation(s)
- Francis M. S. Saraiva
- Trypanosomatids and Vectors Interaction Laboratory, Department of Biochemistry, Roberto Alcantara Gomes Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
| | - Daniela Cosentino-Gomes
- Institute of Medical Biochemistry Leopoldo De Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Job D. F. Inacio
- Tripanosomatide Biochemistry Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro 21040-900, Brazil
| | - Elmo E. Almeida-Amaral
- Tripanosomatide Biochemistry Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro 21040-900, Brazil
| | - Orlando Louzada-Neto
- Laboratory of Toxicology and Molecular Biology, Department of Biochemistry, IBRAG- UERJ, Rio de Janeiro 20511-010, Brazil
| | - Ana Rossini
- Laboratory of Toxicology and Molecular Biology, Department of Biochemistry, IBRAG- UERJ, Rio de Janeiro 20511-010, Brazil
| | - Natália P. Nogueira
- Trypanosomatids and Vectors Interaction Laboratory, Department of Biochemistry, Roberto Alcantara Gomes Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
- National Institute of Science and Technology—Molecular Entomology (INCT-EM), Brasília 70000-000, Brazil
| | - José R. Meyer-Fernandes
- Institute of Medical Biochemistry Leopoldo De Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Marcia C. Paes
- Trypanosomatids and Vectors Interaction Laboratory, Department of Biochemistry, Roberto Alcantara Gomes Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
- National Institute of Science and Technology—Molecular Entomology (INCT-EM), Brasília 70000-000, Brazil
- Correspondence:
| |
Collapse
|
17
|
Mora-Criollo P, Basu R, Qian Y, Costales JA, Guevara-Aguirre J, Grijalva MJ, Kopchick JJ. Growth hormone modulates Trypanosoma cruzi infection in vitro. Growth Horm IGF Res 2022; 64:101460. [PMID: 35490602 DOI: 10.1016/j.ghir.2022.101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 04/11/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Chagas disease (CD) is caused by the protozoan parasite, Trypanosoma cruzi. It affects 7 to 8 million people worldwide and leads to approximately 50,000 deaths per year. In vitro and in vivo studies had demonstrated that Trypanosoma cruziinfection causes an imbalance in the hypothalamic-pituitary-adrenal (HPA) axis that is accompanied by a progressive decrease in growth hormone (GH) and prolactin (PRL) production. In humans, inactivating mutations in the GH receptor gene cause Laron Syndrome (LS), an autosomal recessive disorder. Affected subjects are short, have increased adiposity, decreased insulin-like growth factor-I (IGFI), increased serum GH levels, are highly resistant to diabetes and cancer, and display slow cognitive decline. In addition, CD incidence in these individuals is diminished despite living in highly endemic areas. Consequently, we decided to investigate the in vitro effect of GH/IGF-I on T. cruzi infection. DESIGN We first treated the parasite and/or host cells with different peptide hormones including GH, IGFI, and PRL. Then, we treated cells using different combinations of GH/IGF-I attempting to mimic the GH/IGF-I serum levels observed in LS subjects. RESULTS We found that exogenous GH confers protection against T. cruzi infection. Moreover, this effect is mediated by GH and not IGFI. The combination of relatively high GH (50 ng/ml) and low IGF-I (20 ng/ml), mimicking the hormonal pattern seen in LS individuals, consistently decreased T. cruzi infection in vitro. CONCLUSIONS The combination of relatively high GH and low IGF-I serum levels in LS individuals may be an underlying condition providing partial protection against T. cruzi infection.
Collapse
Affiliation(s)
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Jaime A Costales
- Centro de Investigación para la Salud en América Latina, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Jaime Guevara-Aguirre
- Colegio de ciencias de la salud, Universidad San Francisco de Quito, Cumbaya, Quito, Ecuador
| | - Mario J Grijalva
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH, USA; Centro de Investigación para la Salud en América Latina, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA; Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
18
|
De-Simone SG, Bourguignon SC, Gonçalves PS, Lechuga GC, Provance DW. Metabolic Alteration of Trypanosoma cruzi during Differentiation of Epimastigote to Trypomastigote Forms. Pathogens 2022; 11:pathogens11020268. [PMID: 35215210 PMCID: PMC8879499 DOI: 10.3390/pathogens11020268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/10/2022] Open
Abstract
Intracellular parasites such as Trypanosoma cruzi need to acquire valuable carbon sources from the host cell to replicate. Here, we investigated the energetic metabolism of T. cruzi during metacyclogenesis through the determination of enzymatic activities and quantification by HPLC of glycolytic and Krebs cycle short-chain carboxylic acids. Altered concentrations in pyruvate, acetate, succinate, and glycerate were measured during the growth of epimastigote in the complex medium BHI and their differentiation to trypomastigotes in the chemically defined medium, TAU3AAG. These alterations should represent significant differential metabolic modifications utilized by either form to generate energy. This paper is the first work dealing with the intracellular organic acid concentration measurement in T. cruzi parasites. Although it confirms the previous assumption of the importance of carbohydrate metabolism, it yields an essential improvement in T. cruzi metabolism knowledge.
Collapse
Affiliation(s)
- Salvatore G. De-Simone
- Center for Technological Development in Health (CDTS), FIOCRUZ, National Institute of Science and Technology for Innovation in Neglected Populations Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (P.S.G.); (G.C.L.); (D.W.P.J.)
- Epidemiology and Molecular Systematic Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil
- Cellular and Molecular Biology Department, Biology Institute, Federal Fluminense University, Niterói 24020-141, RJ, Brazil;
- Correspondence:
| | - Saulo C. Bourguignon
- Cellular and Molecular Biology Department, Biology Institute, Federal Fluminense University, Niterói 24020-141, RJ, Brazil;
| | - Priscila S. Gonçalves
- Center for Technological Development in Health (CDTS), FIOCRUZ, National Institute of Science and Technology for Innovation in Neglected Populations Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (P.S.G.); (G.C.L.); (D.W.P.J.)
- Cellular and Molecular Biology Department, Biology Institute, Federal Fluminense University, Niterói 24020-141, RJ, Brazil;
| | - Guilherme C. Lechuga
- Center for Technological Development in Health (CDTS), FIOCRUZ, National Institute of Science and Technology for Innovation in Neglected Populations Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (P.S.G.); (G.C.L.); (D.W.P.J.)
- Epidemiology and Molecular Systematic Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil
| | - David W. Provance
- Center for Technological Development in Health (CDTS), FIOCRUZ, National Institute of Science and Technology for Innovation in Neglected Populations Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (P.S.G.); (G.C.L.); (D.W.P.J.)
- Epidemiology and Molecular Systematic Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, RJ, Brazil
| |
Collapse
|
19
|
The Oxidative Stress and Chronic Inflammatory Process in Chagas Disease: Role of Exosomes and Contributing Genetic Factors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4993452. [PMID: 34976301 PMCID: PMC8718323 DOI: 10.1155/2021/4993452] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/27/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
Chagas disease is a neglected tropical disease caused by the flagellated protozoa Trypanosoma cruzi that affects several million people mainly in Latin American countries. Chagas disease has two phases, which are acute and chronic, both separated by an indeterminate time period in which the infected individual is relatively asymptomatic. The acute phase extends for 40-60 days with atypical and mild symptoms; however, about 30% of the infected patients will develop a symptomatic chronic phase, which is characterized by either cardiac, digestive, neurological, or endocrine problems. Cardiomyopathy is the most important and severe result of Chagas disease, which leads to left ventricular systolic dysfunction, heart failure, and sudden cardiac death. Most deaths are due to heart failure (70%) and sudden death (30%) resulting from cardiomyopathy. During the chronic phase, T. cruzi-infected macrophages respond with the production of proinflammatory cytokines and production of superoxide and nitric oxide by the NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) enzymes, respectively. During the chronic phase, myocardial changes are produced as a result of chronic inflammation, oxidative stress, fibrosis, and cell death. The cellular inflammatory response is mainly the result of activation of the NF-κB-dependent pathway, which activates gene expression of inflammatory cytokines, leading to progressive tissue damage. The persisting production of reactive oxygen species (ROS) is the result of mitochondrial dysfunction in the cardiomyocytes. In this review, we will discuss inflammation and oxidative damage which is produced in the heart during the chronic phase of Chagas disease and recent evidence on the role of macrophages and the production of proinflammatory cytokines during the acute phase and the origin of macrophages/monocytes during the chronic phase of Chagas disease. We will also discuss the contributing factors and mechanisms leading to the chronic inflammation of the cardiac tissue during the chronic phase of the disease as well as the innate and adaptive host immune response. The contribution of genetic factors to the progression of the chronic inflammatory cardiomyopathy of chronic Chagas disease is also discussed. The secreted extracellular vesicles (exosomes) produced for both T. cruzi and infected host cells can play key roles in the host immune response, and those roles are described. Lastly, we describe potential treatments to attenuate the chronic inflammation of the cardiac tissue, designed to improve heart function in chagasic patients.
Collapse
|
20
|
Gachet-Castro C, Freitas-Castro F, Gonzáles-Córdova RA, da Fonseca CK, Gomes MD, Ishikawa-Ankerhold HC, Baqui MMA. Modulation of the Host Nuclear Compartment by Trypanosoma cruzi Uncovers Effects on Host Transcription and Splicing Machinery. Front Cell Infect Microbiol 2021; 11:718028. [PMID: 34737973 PMCID: PMC8560699 DOI: 10.3389/fcimb.2021.718028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/24/2021] [Indexed: 11/22/2022] Open
Abstract
Host manipulation is a common strategy for invading pathogens. Trypanosoma cruzi, the causative agent of Chagas Disease, lives intracellularly within host cells. During infection, parasite-associated modifications occur to the host cell metabolism and morphology. However, little is known about the effect of T. cruzi infection on the host cell nucleus and nuclear functionality. Here, we show that T. cruzi can modulate host transcription and splicing machinery in non-professional phagocytic cells during infection. We found that T. cruzi regulates host RNA polymerase II (RNAPII) in a time-dependent manner, resulting in a drastic decrease in RNAPII activity. Furthermore, host cell ribonucleoproteins associated with mRNA transcription (hnRNPA1 and AB2) are downregulated concurrently. We reasoned that T. cruzi may hijack the host U2AF35 auxiliary factor, a key regulator for RNA processing, as a strategy to affect the splicing machinery activities directly. In support of our hypothesis, we carried out in vivo splicing assays using an adenovirus E1A pre-mRNA splicing reporter, showing that intracellular T. cruzi directly modulates the host cells by appropriating U2AF35. For the first time, our results provide evidence of a complex and intimate molecular relationship between T. cruzi and the host cell nucleus during infection.
Collapse
Affiliation(s)
- Camila Gachet-Castro
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Freitas-Castro
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Raul Alexander Gonzáles-Córdova
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carol Kobori da Fonseca
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Damário Gomes
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Munira Muhammad Abdel Baqui
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
21
|
Liu Z, Ulrich vonBargen R, McCall LI. Central role of metabolism in Trypanosoma cruzi tropism and Chagas disease pathogenesis. Curr Opin Microbiol 2021; 63:204-209. [PMID: 34455304 PMCID: PMC8463485 DOI: 10.1016/j.mib.2021.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 01/17/2023]
Abstract
Chagas disease is a neglected tropical disease caused by Trypanosoma cruzi parasites. During mammalian infection, T. cruzi alternates between an intracellular stage and extracellular stage. T. cruzi adapts its metabolism to this lifestyle, while also reshaping host metabolic pathways. Such host metabolic adaptations compensate for parasite-induced stress, but may promote parasite survival and proliferation. Recent work has demonstrated that metabolism controls parasite tropism and location of Chagas disease symptoms, and regulates whether infection is mild or severe. Such findings have important translational applications with regards to treatment and diagnostic test development, though further research is needed with regards to in vivo parasite metabolic gene expression, relationship between magnitude of local metabolic perturbation, parasite strain and disease location, and host-parasite-microbiota co-metabolism.
Collapse
Affiliation(s)
- Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, 73019, United States; Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, 73019, United States
| | - Rebecca Ulrich vonBargen
- Department of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, United States
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, 73019, United States; Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, 73019, United States; Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, 73019, United States.
| |
Collapse
|
22
|
Gomes MT, Paes-Vieira L, Gomes-Vieira AL, Cosentino-Gomes D, da Silva APP, Giarola NLL, Da Silva D, Sola-Penna M, Galina A, Meyer-Fernandes JR. 3-Bromopyruvate: A new strategy for inhibition of glycolytic enzymes in Leishmania amazonensis. Exp Parasitol 2021; 229:108154. [PMID: 34481863 DOI: 10.1016/j.exppara.2021.108154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/14/2021] [Accepted: 08/28/2021] [Indexed: 11/29/2022]
Abstract
The compound 3-bromopyruvate (3-BrPA) is well-known and studies from several researchers have demonstrated its involvement in tumorigenesis. It is an analogue of pyruvic acid that inhibits ATP synthesis by inhibiting enzymes from the glycolytic pathway and oxidative phosphorylation. In this work, we investigated the effect of 3-BrPA on energy metabolism of L. amazonensis. In order to verify the effect of 3-BrPA on L. amazonensis glycolysis, we measured the activity level of three glycolytic enzymes located at different points of the pathway: (i) glucose kinases, step 1, (ii) glyceraldehyde 3-phosphate dehydrogenase (GAPDH), step 6, and (iii) enolase, step 9. 3-BrPA, in a dose-dependent manner, significantly reduced the activity levels of all the enzymes. In addition, 3-BrPA treatment led to a reduction in the levels of phosphofruto-1-kinase (PFK) protein, suggesting that the mode of action of 3-BrPA involves the downregulation of some glycolytic enzymes. Measurement of ATP levels in promastigotes of L. amazonensis showed a significant reduction in ATP generation. The O2 consumption was also significantly inhibited in promastigotes, confirming the energy depletion effect of 3-BrPA. When 3-BrPA was added to the cells at the beginning of growth cycle, it significantly inhibited L. amazonensis proliferation in a dose-dependent manner. Furthermore, the ability to infect macrophages was reduced by approximately 50% when promastigotes were treated with 3-BrPA. Taken together, these studies corroborate with previous reports which suggest 3-BrPA as a potential drug against pathogenic microorganisms that are reliant on glucose catabolism for ATP supply.
Collapse
Affiliation(s)
- Marta Teixeira Gomes
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA; Laboratório de Bioquímica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Lisvane Paes-Vieira
- Laboratório de Bioquímica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - André Luiz Gomes-Vieira
- Instituto de Química, Departamento de Bioquímica, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Daniela Cosentino-Gomes
- Instituto de Química, Departamento de Bioquímica, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Ana Paula Pereira da Silva
- Instituto de Química, Departamento de Bioquímica, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Naira Ligia Lima Giarola
- Laboratório de Bioquímica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel Da Silva
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio Galina
- Laboratorio de Bioenergética e Fisiologia Mitocondrial, Programa de Bioquímica e Biofísica Celular, Instituto de Bioquímica Medica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Laboratório de Bioquímica Celular, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
23
|
Dumoulin PC, Burleigh BA. Metabolic flexibility in Trypanosoma cruzi amastigotes: implications for persistence and drug sensitivity. Curr Opin Microbiol 2021; 63:244-249. [PMID: 34455305 DOI: 10.1016/j.mib.2021.07.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/18/2022]
Abstract
Throughout their life cycle, parasitic organisms experience a variety of environmental conditions. To ensure persistence and transmission, some protozoan parasites are capable of adjusting their replication or converting to distinct life cycle stages. Trypanosoma cruzi is a 'generalist' parasite that is competent to infect various insect (triatomine) vectors and mammalian hosts. Within the mammalian host, T. cruzi replicates intracellularly as amastigotes and can persist for the lifetime of the host. The persistence of the parasites in tissues can lead to the development of Chagas disease. Recent work has identified growth plasticity and metabolic flexibility as aspects of amastigote biology that are important determinants of persistence in varied growth conditions and under drug pressure. A better understanding of the link between amastigote and host/tissue metabolism will aid in the development of new drugs or therapies that can limit disease pathology.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States.
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| |
Collapse
|
24
|
Medeiros TC, Mehra C, Pernas L. Contact and competition between mitochondria and microbes. Curr Opin Microbiol 2021; 63:189-194. [PMID: 34411806 DOI: 10.1016/j.mib.2021.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 12/01/2022]
Abstract
Invading microbes occupy the host cytosol and take up nutrients on which host organelles are also dependent. Thus, host organelles are poised to interact with intracellular microbes. Despite the essential role of host mitochondria in cellular metabolic homeostasis and in mediating cellular responses to microbial infection, we know little of how these organelles interact with intracellular pathogens, and how such interactions affect disease pathogenesis. Here, we give an overview of the different classes of physical and metabolic interactions reported to occur between mitochondria and eukaryotic pathogens. Investigating the underlying molecular mechanisms and functions of such interactions will reveal novel aspects of infection biology.
Collapse
Affiliation(s)
- Tânia C Medeiros
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Chahat Mehra
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Lena Pernas
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
25
|
Green SB, Lanier RJ, Carey SM, Morgan DR, Gracz H, Sherman J, Rodriguez A, D'Antonio EL. Synthesis, biochemical, and biological evaluation of C2 linkage derivatives of amino sugars, inhibitors of glucokinase from Trypanosoma cruzi. Bioorg Med Chem Lett 2021; 47:128227. [PMID: 34174398 DOI: 10.1016/j.bmcl.2021.128227] [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: 04/29/2021] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 11/25/2022]
Abstract
Eighteen amino sugar analogues were screened against Trypanosoma cruzi glucokinase (TcGlcK), a potential drug-target of the protozoan parasite in order to assess for viable enzyme inhibition. The analogues were divided into three amino sugar scaffolds that included d-glucosamine (d-GlcN), d-mannosamine (d-ManN), and d-galactosamine (d-GalN); moreover, all but one of these compounds were novel. TcGlcK is an important metabolic enzyme that has a role in producing G6P for glycolysis and the pentose phosphate pathway (PPP). The inhibition of these pathways via glucose kinases (i.e., glucokinase and hexokinase) appears to be a strategic approach for drug discovery. Glucose kinases phosphorylate d-glucose with co-substrate ATP to yield G6P and the formed G6P enters both pathways for catabolism. The compound screen revealed five on-target confirmed inhibitors that were all from the d-GlcN series, such as compounds 1, 2, 4, 5, and 6. Four of these compounds were strong TcGlcK inhibitors (1, 2, 4, and 6) since they were found to have micromolar inhibitory constant (Ki) values around 20 μM. Three of the on-target confirmed inhibitors (1, 5, and 6) revealed notable in vitro anti-T. cruzi activity with IC50 values being less than 50 μM. Compound 1 was benzoyl glucosamine (BENZ-GlcN), a known TcGlcK inhibitor that was the starting point for the design of the compounds in this study; in addition, TcGlcK - compound 1 inhibition properties were previously determined [D'Antonio, E. L. et al. (2015) Mol. Biochem. Parasitol. 204, 64-76]. As such, compounds 5 and 6 were further evaluated biochemically, where formal Ki values were determined as well as their mode of TcGlcK inhibition. The Ki values determined for compounds 5 and 6 were 107 ± 4 μM and 15.2 ± 3.3 μM, respectively, and both of these compounds exhibited the competitive inhibition mode.
Collapse
Affiliation(s)
- Scott B Green
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Robert J Lanier
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Shane M Carey
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | | | - Hanna Gracz
- NMRService LLC, Raleigh, NC 27612, USA; Department of Molecular and Structural Biochemistry, North Carolina State University, 128 Polk Hall, Raleigh, NC 27695, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, 430 East 29(th) Street, New York, NY 10016, USA
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, 430 East 29(th) Street, New York, NY 10016, USA
| | - Edward L D'Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA.
| |
Collapse
|
26
|
Pei G, Dorhoi A. NOD-Like Receptors: Guards of Cellular Homeostasis Perturbation during Infection. Int J Mol Sci 2021; 22:ijms22136714. [PMID: 34201509 PMCID: PMC8268748 DOI: 10.3390/ijms22136714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 12/30/2022] Open
Abstract
The innate immune system relies on families of pattern recognition receptors (PRRs) that detect distinct conserved molecular motifs from microbes to initiate antimicrobial responses. Activation of PRRs triggers a series of signaling cascades, leading to the release of pro-inflammatory cytokines, chemokines and antimicrobials, thereby contributing to the early host defense against microbes and regulating adaptive immunity. Additionally, PRRs can detect perturbation of cellular homeostasis caused by pathogens and fine-tune the immune responses. Among PRRs, nucleotide binding oligomerization domain (NOD)-like receptors (NLRs) have attracted particular interest in the context of cellular stress-induced inflammation during infection. Recently, mechanistic insights into the monitoring of cellular homeostasis perturbation by NLRs have been provided. We summarize the current knowledge about the disruption of cellular homeostasis by pathogens and focus on NLRs as innate immune sensors for its detection. We highlight the mechanisms employed by various pathogens to elicit cytoskeleton disruption, organelle stress as well as protein translation block, point out exemplary NLRs that guard cellular homeostasis during infection and introduce the concept of stress-associated molecular patterns (SAMPs). We postulate that integration of information about microbial patterns, danger signals, and SAMPs enables the innate immune system with adequate plasticity and precision in elaborating responses to microbes of variable virulence.
Collapse
Affiliation(s)
- Gang Pei
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany
- Correspondence: (G.P.); (A.D.)
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, 17489 Greifswald, Germany
- Correspondence: (G.P.); (A.D.)
| |
Collapse
|
27
|
Alcantara CL, de Souza W, Cunha E Silva NL. The cytostome-cytopharynx complex of intracellular and extracellular amastigotes of Trypanosoma cruzi exhibit structural and functional differences. Cell Microbiol 2021; 23:e13346. [PMID: 33900003 DOI: 10.1111/cmi.13346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 01/01/2023]
Abstract
Endocytosis in Trypanosoma cruzi is mainly performed through a specialised membrane domain called cytostome-cytopharynx complex. Its ultrastructure and dynamics in endocytosis are well characterized in epimastigotes, being absent in trypomastigotes, that lack endocytic activity. Intracellular amastigotes also possess a cytostome-cytopharynx but participation in endocytosis of these forms is not clear. Extracellular amastigotes can be obtained from the supernatant of infected cells or in vitro amastigogenesis. These amastigotes share biochemical and morphological features with intracellular amastigotes but retain trypomastigote's ability to establish infection. We analysed and compared the ultrastructure of the cytostome-cytopharynx complex of intracellular amastigotes and extracellular amastigotes using high-resolution tridimensional electron microscopy techniques. We compared the endocytic ability of intracellular amastigotes, obtained through host cell lysis, with that of extracellular amastigotes. Intracellular amastigotes showed a cytostome-cytopharynx complex similar to epimastigotes'. However, after isolation, the complex undergoes ultrastructural modifications that progressively took to an impairment of endocytosis. Extracellular amastigotes do not possess a cytostome-cytopharynx complex nor the ability to endocytose. Those observations highlight morpho functional differences between intra and extracellular amastigotes regarding an important structure related to cell metabolism. TAKE AWAYS: T. cruzi intracellular amastigotes endocytose through the cytostome-cytopharynx complex. The cytostome-cytopharynx complex of intracellular amastigotes is ultrastructurally similar to the epimastigote. Intracellular amastigotes, once outside the host cell, disassembles the cytostome-cytopharynx membrane domain. Extracellular amastigotes do not possess a cytostome-cytopharynx either the ability to endocytose.
Collapse
Affiliation(s)
- Carolina L Alcantara
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Núcleo de Biologia Estrutural e Bioimagens (CENABIO)-Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Biomagens (INBEB), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Núcleo de Biologia Estrutural e Bioimagens (CENABIO)-Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Biomagens (INBEB), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Narcisa L Cunha E Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.,Núcleo de Biologia Estrutural e Bioimagens (CENABIO)-Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Biomagens (INBEB), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Lander N, Chiurillo MA, Docampo R. Signaling pathways involved in environmental sensing in Trypanosoma cruzi. Mol Microbiol 2021; 115:819-828. [PMID: 33034088 PMCID: PMC8032824 DOI: 10.1111/mmi.14621] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/18/2022]
Abstract
Trypanosoma cruzi is a unicellular parasite and the etiologic agent of Chagas disease. The parasite has a digenetic life cycle alternating between mammalian and insect hosts, where it faces a variety of environmental conditions to which it must adapt in order to survive. The adaptation to these changes is mediated by signaling pathways that coordinate the cellular responses to the new environmental settings. Major environmental changes include temperature, nutrient availability, ionic composition, pH, osmolarity, oxidative stress, contact with host cells and tissues, host immune response, and intracellular life. Some of the signaling pathways and second messengers potentially involved in the response to these changes have been elucidated in recent years and will be the subject of this review.
Collapse
Affiliation(s)
- Noelia Lander
- Center for Tropical and Emerging Global Diseases, and Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Miguel A. Chiurillo
- Center for Tropical and Emerging Global Diseases, and Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Roberto Docampo
- Center for Tropical and Emerging Global Diseases, and Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
29
|
Mitochondrial Pyruvate Carrier Subunits Are Essential for Pyruvate-Driven Respiration, Infectivity, and Intracellular Replication of Trypanosoma cruzi. mBio 2021; 12:mBio.00540-21. [PMID: 33824204 PMCID: PMC8092248 DOI: 10.1128/mbio.00540-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease. Pyruvate is the end product of glycolysis, and its transport into the mitochondrion is mediated by the mitochondrial pyruvate carrier (MPC) subunits. Pyruvate is the final metabolite of glycolysis and can be converted into acetyl coenzyme A (acetyl-CoA) in mitochondria, where it is used as the substrate for the tricarboxylic acid cycle. Pyruvate availability in mitochondria depends on its active transport through the heterocomplex formed by the mitochondrial pyruvate carriers 1 and 2 (MPC1/MPC2). We report here studies on MPC1/MPC2 of Trypanosoma cruzi, the etiologic agent of Chagas disease. Endogenous tagging of T. cruziMPC1 (TcMPC1) and TcMPC2 with 3×c-Myc showed that both encoded proteins colocalize with MitoTracker to the mitochondria of epimastigotes. Individual knockout (KO) of TcMPC1 and TcMPC2 genes using CRISPR/Cas9 was confirmed by PCR and Southern blot analyses. Digitonin-permeabilized TcMPC1-KO and TcMPC2-KO epimastigotes showed reduced O2 consumption rates when pyruvate, but not succinate, was used as the mitochondrial substrate, while α-ketoglutarate increased their O2 consumption rates due to an increase in α-ketoglutarate dehydrogenase activity. Defective mitochondrial pyruvate import resulted in decreased Ca2+ uptake. The inhibitors UK5099 and malonate impaired pyruvate-driven oxygen consumption in permeabilized control cells. Inhibition of succinate dehydrogenase by malonate indicated that pyruvate needs to be converted into succinate to increase respiration. TcMPC1-KO and TcMPC2-KO epimastigotes showed little growth differences in standard or low-glucose culture medium. However, the ability of trypomastigotes to infect tissue culture cells and replicate as intracellular amastigotes was decreased in TcMPC-KOs. Overall, T. cruzi MPC1 and MPC2 are essential for cellular respiration in the presence of pyruvate, invasion of host cells, and replication of amastigotes.
Collapse
|
30
|
Salem DA, Salem NA, Hendawy SR. Association between Toxoplasma gondii infection and metabolic syndrome in obese adolescents: A possible immune-metabolic link. Parasitol Int 2021; 83:102343. [PMID: 33831579 DOI: 10.1016/j.parint.2021.102343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/27/2021] [Accepted: 04/01/2021] [Indexed: 01/22/2023]
Abstract
Background Toxoplasmosis as a global disease is considered as a triggering factor responsible for development of several clinical diseases. However, Toxoplasma gondii (T. gondii) is an understudied parasite of potential interest in obesity research. The current study aimed to explore the role of latent T. gondii infection in the pathogenesis of metabolic syndrome (MetS) in obese adolescents through studying the relationship between serum interferon-gamma [IFN-γ] and serum chemerin in context of MetS components. Methods Eighty-three obese adolescents were serologically screened for T. gondii-IgG antibodies and compared to 35 age-matched healthy T. gondii-seronegative controls. Participants were evaluated for anthropometric measurements, total-fat mass [FM], trunk-FM, serum lipid profile, IFN-γ, and chemerin levels. Homeostatic Model Assessment of insulin resistance (HOMA-IR) was calculated. Results The prevalence of MetS was significantly higher within obese T. gondii-seropositive group compared to obese T. gondii-seronegative group (P = 0.033). Seropositive obese MetS group displayed significantly higher trunk-FM, HOMA-IR, chemerin, and IFN-γ compared to seronegative obese MetS group. Serum chemerin and IFN-γ were strongly correlated (P < 0.001) and were positively correlated with BMI, WC, total-FM, trunk-FM, HOMA-IR, cholesterol, triglycerides and negatively correlated with HDLC. HOMA-IR was a common predictor for serum chemerin (P = 0.030) and IFN-γ (P < 0.001). Conclusions The study results suggest that T. gondii infection may exert an immune-metabolic effect that may have a potential role in the development of MetS among obese adolescents.
Collapse
Affiliation(s)
- Doaa A Salem
- Medical Parasitology Department, Faculty of Medicine, Mansoura University, Egypt.
| | - Nanees A Salem
- Department of Pediatrics, Pediatric Endocrinology Unit, Faculty of Medicine, Mansoura University, Egypt
| | - Shimaa R Hendawy
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
31
|
Parab AR, McCall LI. Tryp-ing Up Metabolism: Role of Metabolic Adaptations in Kinetoplastid Disease Pathogenesis. Infect Immun 2021; 89:e00644-20. [PMID: 33526564 PMCID: PMC8090971 DOI: 10.1128/iai.00644-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Today, more than a billion people-one-sixth of the world's population-are suffering from neglected tropical diseases. Human African trypanosomiasis, Chagas disease, and leishmaniasis are neglected tropical diseases caused by protozoan parasites belonging to the genera Trypanosoma and Leishmania About half a million people living in tropical and subtropical regions of the world are at risk of contracting one of these three infections. Kinetoplastids have complex life cycles with different morphologies and unique physiological requirements at each life cycle stage. This review covers the latest findings on metabolic pathways impacting disease pathogenesis of kinetoplastids within the mammalian host. Nutrient availability is a key factor shaping in vivo parasite metabolism; thus, kinetoplastids display significant metabolic flexibility. Proteomic and transcriptomic profiles show that intracellular trypanosomatids are able to switch to an energy-efficient metabolism within the mammalian host system. Host metabolic changes can also favor parasite persistence, and contribute to symptom development, in a location-specific fashion. Ultimately, targeted and untargeted metabolomics studies have been a valuable approach to elucidate the specific biochemical pathways affected by infection within the host, leading to translational drug development and diagnostic insights.
Collapse
Affiliation(s)
- Adwaita R Parab
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
32
|
Libisch MG, Rego N, Robello C. Transcriptional Studies on Trypanosoma cruzi - Host Cell Interactions: A Complex Puzzle of Variables. Front Cell Infect Microbiol 2021; 11:692134. [PMID: 34222052 PMCID: PMC8248493 DOI: 10.3389/fcimb.2021.692134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/26/2021] [Indexed: 01/05/2023] Open
Abstract
Chagas Disease, caused by the protozoan parasite Trypanosoma cruzi, affects nearly eight million people in the world. T. cruzi is a complex taxon represented by different strains with particular characteristics, and it has the ability to infect and interact with almost any nucleated cell. The T. cruzi-host cell interactions will trigger molecular signaling cascades in the host cell that will depend on the particular cell type and T. cruzi strain, and also on many different experimental variables. In this review we collect data from multiple transcriptomic and functional studies performed in different infection models, in order to highlight key differences between works that in our opinion should be addressed when comparing and discussing results. In particular, we focus on changes in the respiratory chain and oxidative phosphorylation of host cells in response to infection, which depends on the experimental model of T. cruzi infection. Finally, we also discuss host cell responses which reiterate independently of the strain, cell type and experimental conditions.
Collapse
Affiliation(s)
- María Gabriela Libisch
- Laboratorio de Interacciones Hospedero Patógeno-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Natalia Rego
- Unidad de Bioinformática, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero Patógeno-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- *Correspondence: Carlos Robello,
| |
Collapse
|
33
|
Dumoulin PC, Vollrath J, Tomko SS, Wang JX, Burleigh B. Glutamine metabolism modulates azole susceptibility in Trypanosoma cruzi amastigotes. eLife 2020; 9:60226. [PMID: 33258448 PMCID: PMC7707839 DOI: 10.7554/elife.60226] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/17/2020] [Indexed: 12/27/2022] Open
Abstract
The mechanisms underlying resistance of the Chagas disease parasite, Trypanosoma cruzi, to current therapies are not well understood, including the role of metabolic heterogeneity. We found that limiting exogenous glutamine protects actively dividing amastigotes from ergosterol biosynthesis inhibitors (azoles), independent of parasite growth rate. The antiparasitic properties of azoles are derived from inhibition of lanosterol 14α-demethylase (CYP51) in the endogenous sterol synthesis pathway. We find that carbons from 13C-glutamine feed into amastigote sterols and into metabolic intermediates that accumulate upon CYP51 inhibition. Incorporation of 13C-glutamine into endogenously synthesized sterols is increased with BPTES treatment, an inhibitor of host glutamine metabolism that sensitizes amastigotes to azoles. Similarly, amastigotes are re-sensitized to azoles following addition of metabolites upstream of CYP51, raising the possibility that flux through the sterol synthesis pathway is a determinant of sensitivity to azoles and highlighting the potential role for metabolic heterogeneity in recalcitrant T. cruzi infection.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Joshua Vollrath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Institute for Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Sheena Shah Tomko
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Jennifer X Wang
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, United States
| | - Barbara Burleigh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States
| |
Collapse
|
34
|
Shiratsubaki IS, Fang X, Souza ROO, Palsson BO, Silber AM, Siqueira-Neto JL. Genome-scale metabolic models highlight stage-specific differences in essential metabolic pathways in Trypanosoma cruzi. PLoS Negl Trop Dis 2020; 14:e0008728. [PMID: 33021977 PMCID: PMC7567352 DOI: 10.1371/journal.pntd.0008728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 10/16/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Chagas disease is a neglected tropical disease and a leading cause of heart failure in Latin America caused by a protozoan called Trypanosoma cruzi. This parasite presents a complex multi-stage life cycle. Anti-Chagas drugs currently available are limited to benznidazole and nifurtimox, both with severe side effects. Thus, there is a need for alternative and more efficient drugs. Genome-scale metabolic models (GEMs) can accurately predict metabolic capabilities and aid in drug discovery in metabolic genes. This work developed an extended GEM, hereafter referred to as iIS312, of the published and validated T. cruzi core metabolism model. From iIS312, we then built three stage-specific models through transcriptomics data integration, and showed that epimastigotes present the most active metabolism among the stages (see S1-S4 GEMs). Stage-specific models predicted significant metabolic differences among stages, including variations in flux distribution in core metabolism. Moreover, the gene essentiality predictions suggest potential drug targets, among which some have been previously proven lethal, including glutamate dehydrogenase, glucokinase and hexokinase. To validate the models, we measured the activity of enzymes in the core metabolism of the parasite at different stages, and showed the results were consistent with model predictions. Our results represent a potential step forward towards the improvement of Chagas disease treatment. To our knowledge, these stage-specific models are the first GEMs built for the stages Amastigote and Trypomastigote. This work is also the first to present an in silico GEM comparison among different stages in the T. cruzi life cycle.
Collapse
Affiliation(s)
- Isabel S Shiratsubaki
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, United States of America
- Department of Bioengineering, UC San Diego, La Jolla, California, United States of America
| | - Xin Fang
- Department of Bioengineering, UC San Diego, La Jolla, California, United States of America
| | - Rodolpho O O Souza
- Laboratory of Biochemistry of Tryps - LaBTryps, Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Bernhard O Palsson
- Department of Bioengineering, UC San Diego, La Jolla, California, United States of America
- Department of Pediatrics, UC San Diego, La Jolla, California, United States of America
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Ariel M Silber
- Laboratory of Biochemistry of Tryps - LaBTryps, Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, United States of America
| |
Collapse
|
35
|
Repurposing bioenergetic modulators against protozoan parasites responsible for tropical diseases. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:17-27. [PMID: 32829099 PMCID: PMC7452664 DOI: 10.1016/j.ijpddr.2020.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/30/2022]
Abstract
Malaria, leishmaniasis and trypanosomiasis are arthropod-borne, parasitic diseases that constitute a major global health problem. They are generally found in developing countries, where lack of access to preventive tools and treatment hinders their management. Because these parasites share an increased demand on glucose consumption with most cancer cells, six compounds used in anti-tumoral research were selected to be tested as antiparasitic agents in in vitro models of Leishmania infantum, Trypanosoma brucei, T. cruzi, and Plasmodium falciparum: dichloroacetic acid (DCA), 3-bromopyruvic acid (3BP), 2-deoxy-D-glucose (2DG), lonidamine (LND), metformin (MET), and sirolimus (SIR). No parasite-killing activity was found in L. infantum promastigotes, whereas DCA and 3BP reduced the burden of intra-macrophagic amastigotes. For T. brucei all selected compounds, but 2DG, decreased parasite survival. DCA, 2DG, LND and MET showed parasite-killing activity in T. cruzi. Finally, anti-plasmodial activity was found for DCA, 2DG, LND, MET and SIR. These results reinforce the hypothesis that drugs with proven efficacy in the treatment of cancer by interfering with ATP production, proliferation, and survival cell strategies might be useful in treating threatening parasitic diseases and provide new opportunities for their repurposing. Parasitic diseases are prevalent among the poorest of the poor. Some parasitic protists degrade glucose into CO2 even aerobically making this a target. Degrading glucose into CO2 (Warburg effect) is also characteristic for cancer cells. Repurposing cancer glycolysis blockers may provide cost-effective treatments for the poorest.
Collapse
|
36
|
Fernandes PM, Kinkead J, McNae IW, Vásquez-Valdivieso M, Wear MA, Michels PAM, Walkinshaw MD. Kinetic and structural studies of Trypanosoma and Leishmania phosphofructokinases show evolutionary divergence and identify AMP as a switch regulating glycolysis versus gluconeogenesis. FEBS J 2020; 287:2847-2861. [PMID: 31838765 PMCID: PMC7383607 DOI: 10.1111/febs.15177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/13/2019] [Accepted: 12/12/2019] [Indexed: 11/30/2022]
Abstract
Trypanosomatids possess glycosome organelles that contain much of the glycolytic machinery, including phosphofructokinase (PFK). We present kinetic and structural data for PFK from three human pathogenic trypanosomatids, illustrating intriguing differences that may reflect evolutionary adaptations to differing ecological niches. The activity of Leishmania PFK – to a much larger extent than Trypanosoma PFK – is reliant on AMP for activity regulation, with 1 mm AMP increasing the L. infantum PFK (LiPFK) kcat/K0.5F6P value by 10‐fold, compared to only a 1.3‐ and 1.4‐fold increase for T. cruzi and T. brucei PFK, respectively. We also show that Leishmania PFK melts at a significantly lower (> 15 °C) temperature than Trypanosoma PFKs and that addition of either AMP or ATP results in a marked stabilization of the protein. Sequence comparisons of Trypanosoma spp. and Leishmania spp. show that divergence of the two genera involved amino acid substitutions that occur in the enzyme’s ‘reaching arms’ and ‘embracing arms’ that determine tetramer stability. The dramatic effects of AMP on Leishmania activity compared with the Trypanosoma PFKs may be explained by differences between the T‐to‐R equilibria for the two families, with the low‐melting Leishmania PFK favouring the flexible inactive T‐state in the absence of AMP. Sequence comparisons along with the enzymatic and structural data presented here also suggest there was a loss of AMP‐dependent regulation in Trypanosoma species rather than gain of this characteristic in Leishmania species and that AMP acts as a key regulator in Leishmania governing the balance between glycolysis and gluconeogenesis.
Collapse
Affiliation(s)
- Peter M Fernandes
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - James Kinkead
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Iain W McNae
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Monserrat Vásquez-Valdivieso
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Martin A Wear
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Paul A M Michels
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Malcolm D Walkinshaw
- Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Dumoulin PC, Burleigh BA. Methods for the Investigation of Trypanosoma cruzi Amastigote Proliferation in Mammalian Host Cells. Methods Mol Biol 2020; 2116:535-554. [PMID: 32221941 DOI: 10.1007/978-1-0716-0294-2_32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In its mammalian host, the kinetoplastid protozoan parasite, Trypanosoma cruzi, is obliged to establish intracellular residence in order to replicate. This parasite can infect and replicate within a diverse array of cell and tissue types across many mammalian host species. The establishment of quantitative assays to assess the replicative capacity of intracellular T. cruzi amastigotes under different conditions is a critical facet to understanding this host-pathogen interaction. Several complementary methods are outlined here. Their strengths and deficiencies in quantifying intracellular amastigote growth and death are discussed. We describe three assays to assess growth/replication. (1) A high throughput multiplexed plate-based assay that quantifies both host cell and parasite abundance. This method allows for the rapid and simultaneous screening of many conditions (e.g., small molecule inhibitors, the impact of host gene knockdown or of altered environmental parameters). (2) Simple fluorescence microscopy-based enumeration of amastigotes within host cells and (3) flow cytometry-based quantification of amastigote proliferation following isolation from host cells. Each approach has advantages but none of these can assess lethal outcomes in a quantitative manner. For this, we describe a clonal outgrowth assay that identifies the proportion of parasites that succumb to a defined exposure. Even using these assays, it can be challenging to differentiate between direct (targeting the parasite) and/or indirect (targeting the host) effects of a given treatment on amastigote growth. Therefore, we also outline a method of purification of intracellular amastigotes that allows for downstream biochemical and metabolic investigations specifically on the isolated amastigote.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
38
|
Cordeiro AT. NADPH Producing Enzymes as Promising Drug Targets for Chagas Disease. Curr Med Chem 2019; 26:6564-6571. [PMID: 30306853 DOI: 10.2174/0929867325666181009152844] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/10/2018] [Accepted: 09/19/2018] [Indexed: 11/22/2022]
Abstract
Reduced Nicotinamide Adenine Dinucleotide Phosphate (NADPH) is a cofactor used in different anabolic reactions, such as lipid and nucleic acid synthesis, and for oxidative stress defense. NADPH is essential for parasite growth and viability. In trypanosomatid parasites, NADPH is supplied by the oxidative branch of the pentose phosphate pathway and by enzymes associated with the citric acid cycle. The present article will review recent achievements that suggest glucose-6-phosphate dehydrogenase and the cytosolic isoform of the malic enzyme as promising drug targets for the discovery of new drugs against Trypanosoma cruzi and T. brucei. Topics involving an alternative strategy in accelerating T. cruzi drug-target validation and the concept of drug-target classification will also be revisited.
Collapse
Affiliation(s)
- Artur T Cordeiro
- Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas- SP, Brazil
| |
Collapse
|
39
|
Mesías AC, Garg NJ, Zago MP. Redox Balance Keepers and Possible Cell Functions Managed by Redox Homeostasis in Trypanosoma cruzi. Front Cell Infect Microbiol 2019; 9:435. [PMID: 31921709 PMCID: PMC6932984 DOI: 10.3389/fcimb.2019.00435] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
The toxicity of oxygen and nitrogen reactive species appears to be merely the tip of the iceberg in the world of redox homeostasis. Now, oxidative stress can be seen as a two-sided process; at high concentrations, it causes damage to biomolecules, and thus, trypanosomes have evolved a strong antioxidant defense system to cope with these stressors. At low concentrations, oxidants are essential for cell signaling, and in fact, the oxidants/antioxidants balance may be able to trigger different cell fates. In this comprehensive review, we discuss the current knowledge of the oxidant environment experienced by T. cruzi along the different phases of its life cycle, and the molecular tools exploited by this pathogen to deal with oxidative stress, for better or worse. Further, we discuss the possible redox-regulated processes that could be governed by this oxidative context. Most of the current research has addressed the importance of the trypanosomes' antioxidant network based on its detox activity of harmful species; however, new efforts are necessary to highlight other functions of this network and the mechanisms underlying the fine regulation of the defense machinery, as this represents a master key to hinder crucial pathogen functions. Understanding the relevance of this balance keeper program in parasite biology will give us new perspectives to delineate improved treatment strategies.
Collapse
Affiliation(s)
- Andrea C. Mesías
- Instituto de Patología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad Nacional de Salta, Salta, Argentina
| | - Nisha J. Garg
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| | - M. Paola Zago
- Instituto de Patología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad Nacional de Salta, Salta, Argentina
| |
Collapse
|
40
|
Ty MC, Loke P, Alberola J, Rodriguez A, Rodriguez-Cortes A. Immuno-metabolic profile of human macrophages after Leishmania and Trypanosoma cruzi infection. PLoS One 2019; 14:e0225588. [PMID: 31841511 PMCID: PMC6913957 DOI: 10.1371/journal.pone.0225588] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Macrophages can reprogram their metabolism in response to the surrounding stimuli, which affects their capacity to kill intracellular pathogens. We have investigated the metabolic and immune status of human macrophages after infection with the intracellular trypanosomatid parasites Leishmania donovani, L. amazonensis and T. cruzi and their capacity to respond to a classical polarizing stimulus (LPS and IFN-γ). We found that macrophages infected with Leishmania preferentially upregulate oxidative phosphorylation, which could be contributed by both host cell and parasite, while T. cruzi infection did not significantly increase glycolysis or oxidative phosphorylation. Leishmania and T. cruzi infect macrophages without triggering a strong inflammatory cytokine response, but infection does not prevent a potent response to LPS and IFN-γ. Infection appears to prime macrophages, since the cytokine response to activation with LPS and IFN-γ is more intense in infected macrophages compared to uninfected ones. Metabolic polarization in macrophages can influence infection and immune evasion of these parasites since preventing macrophage cytokine responses would help parasites to establish a persistent infection. However, macrophages remain responsive to classical inflammatory stimuli and could still trigger inflammatory cytokine secretion by macrophages.
Collapse
Affiliation(s)
- Maureen C. Ty
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
| | - P’ng Loke
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
| | - Jordi Alberola
- Dept Farmacologia, Toxicologia i Terapeutica, Facultat de Veterinaria, Edifici V, Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Ana Rodriguez
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
- * E-mail:
| | - Alheli Rodriguez-Cortes
- New York University School of Medicine, Department of Microbiology, New York, NY, United States of America
- Dept Farmacologia, Toxicologia i Terapeutica, Facultat de Veterinaria, Edifici V, Universitat Autonoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
41
|
Hennig K, Abi-Ghanem J, Bunescu A, Meniche X, Biliaut E, Ouattara AD, Lewis MD, Kelly JM, Braillard S, Courtemanche G, Chatelain E, Béquet F. Metabolomics, lipidomics and proteomics profiling of myoblasts infected with Trypanosoma cruzi after treatment with different drugs against Chagas disease. Metabolomics 2019; 15:117. [PMID: 31440849 DOI: 10.1007/s11306-019-1583-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/17/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Chagas disease, the most important parasitic infection in Latin America, is caused by the intracellular protozoan Trypanosoma cruzi. To treat this disease, only two nitroheterocyclic compounds with toxic side effects exist and frequent treatment failures are reported. Hence there is an urgent need to develop new drugs. Recently, metabolomics has become an efficient and cost-effective strategy for dissecting drug mode of action, which has been applied to bacteria as well as parasites, such as different Trypanosome species and forms. OBJECTIVES We assessed if the metabolomics approach can be applied to study drug action of the intracellular amastigote form of T. cruzi in a parasite-host cell system. METHODS We applied a metabolic fingerprinting approach (DI-MS and NMR) to evaluate metabolic changes induced by six different (candidate) drugs in a parasite-host cell system. In a second part of our study, we analyzed the impact of two drugs on polar metabolites, lipid and proteins to evaluate if affected pathways can be identified. RESULTS Metabolic signatures, obtained by the fingerprinting approach, resulted in three different clusters. Two can be explained by already known of mode actions, whereas the three experimental drugs formed a separate cluster. Significant changes induced by drug action were observed in all the three metabolic fractions (polar metabolites, lipids and proteins). We identified a general impact on the TCA cycle, but no specific pathways could be attributed to drug action, which might be caused by a high percentage of common metabolome between a eukaryotic host cell and a eukaryotic parasite. Additionally, ion suppression effects due to differences in abundance between host cells and parasites may have occurred. CONCLUSION We validated the metabolic fingerprinting approach to a complex host-cell parasite system. This technique can potentially be applied in the early stage of drug discovery and could help to prioritize early leads or reconfirmed hits for further development.
Collapse
Affiliation(s)
- K Hennig
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - J Abi-Ghanem
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - A Bunescu
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - X Meniche
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - E Biliaut
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - A D Ouattara
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France
| | - M D Lewis
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - J M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - S Braillard
- Drugs for Neglected Diseases Initiative (DNDi), 15 Chemin Louis-Dunant, 1202, Geneva, Switzerland
| | | | - E Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), 15 Chemin Louis-Dunant, 1202, Geneva, Switzerland
| | - F Béquet
- BIOASTER, 40 avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
42
|
Abstract
It is generally regarded that the progression of an infection within host macrophages is the consequence of a failed immune response. However, recent appreciation of macrophage heterogeneity, with respect to both development and metabolism, indicates that the reality is more complex. Different lineages of tissue-resident macrophages respond divergently to microbial, environmental and immunological stimuli. The emerging picture that the developmental origin of macrophages determines their responses to immune stimulation and to infection stresses the importance of in vivo infection models. Recent investigations into the metabolism of infecting microorganisms and host macrophages indicate that their metabolic interface can be a major determinant of pathogen growth or containment. This Review focuses on the integration of data from existing studies, the identification of challenges in generating and interpreting data from ongoing studies and a discussion of the technologies and tools that are required to best address future questions in the field.
Collapse
Affiliation(s)
- David G Russell
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Lu Huang
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Brian C VanderVen
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
43
|
Takagi Y, Akutsu Y, Doi M, Furukawa K. Utilization of proliferable extracellular amastigotes for transient gene expression, drug sensitivity assay, and CRISPR/Cas9-mediated gene knockout in Trypanosoma cruzi. PLoS Negl Trop Dis 2019; 13:e0007088. [PMID: 30640901 PMCID: PMC6347291 DOI: 10.1371/journal.pntd.0007088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 01/25/2019] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Trypanosoma cruzi has three distinct life cycle stages; epimastigote, trypomastigote, and amastigote. Amastigote is the replication stage in host mammalian cells, hence this stage of parasite has clinical significance in drug development research. Presence of extracellular amastigotes (EA) and their infection capability have been known for some decades. Here, we demonstrate that EA can be utilized as an axenic culture to aid in stage-specific study of T. cruzi. Amastigote-like property of axenic amastigote can be sustained in LIT medium at 37°C at least for 1 week, judging from their morphology, amastigote-specific UTR-regulated GFP expression, and stage-specific expression of selected endogenous genes. Inhibitory effect of benznidazole and nifurtimox on axenic amastigotes was comparable to that on intracellular amastigotes. Exogenous nucleic acids can be transfected into EA via conventional electroporation, and selective marker could be utilized for enrichment of transfectants. We also demonstrate that CRISPR/Cas9-mediated gene knockout can be performed in EA. Essentiality of the target gene can be evaluated by the growth capability of the knockout EA, either by continuation of axenic culturing or by host infection and following replication as intracellular amastigotes. By taking advantage of the accessibility and sturdiness of EA, we can potentially expand our experimental freedom in studying amastigote stage of T. cruzi.
Collapse
Affiliation(s)
- Yuko Takagi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
- * E-mail:
| | - Yukie Akutsu
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Koji Furukawa
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| |
Collapse
|
44
|
Legionella pneumophila Is Directly Sensitive to 2-Deoxyglucose-Phosphate via Its UhpC Transporter but Is Indifferent to Shifts in Host Cell Glycolytic Metabolism. J Bacteriol 2018; 200:JB.00176-18. [PMID: 29784886 DOI: 10.1128/jb.00176-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptor (TLR) stimulation induces a pronounced shift to increased glycolytic metabolism in mammalian macrophages. We observed that bone marrow-derived macrophages (BMMs) increase glycolysis in response to infection with Legionella pneumophila, but the role of host macrophage glycolysis in terms of intracellular L. pneumophila replication is not currently understood. Treatment with 2-deoxyglucose (2DG) blocks L. pneumophila replication in mammalian macrophages but has no effect on bacteria grown in broth. In addition, we found that 2DG had no effect on bacteria grown in amoebae. We used a serial enrichment strategy to reveal that the effect of 2DG on L. pneumophila in macrophages requires the L. pneumophila hexose-phosphate transporter UhpC. Experiments with UhpC-deficient L. pneumophila revealed that mutant bacteria are also resistant to growth inhibition following treatment with phosphorylated 2DG in broth, suggesting that the inhibitory effect of 2DG on L. pneumophila in mammalian cells requires 2DG phosphorylation. UhpC-deficient L. pneumophila replicates without a growth defect in BMMs and protozoan host cells and also replicates without a growth defect in BMMs treated with 2DG. Our data indicate that neither TLR signaling-dependent increased macrophage glycolysis nor inhibition of macrophage glycolysis has a substantial effect on intracellular L. pneumophila replication. These results are consistent with the view that L. pneumophila can employ diverse metabolic strategies to exploit its host cells.IMPORTANCE We explored the relationship between macrophage glycolysis and replication of an intracellular bacterial pathogen, Legionella pneumophila Previous studies demonstrated that a glycolysis inhibitor, 2-deoxyglucose (2DG), blocks replication of L. pneumophila during infection of macrophages, leading to speculation that L. pneumophila may exploit macrophage glycolysis. We isolated L. pneumophila mutants resistant to the inhibitory effect of 2DG in macrophages, identifying a L. pneumophila hexose-phosphate transporter, UhpC, that is required for bacterial sensitivity to 2DG during infection. Our results reveal how a bacterial transporter mediates the direct antimicrobial effect of a toxic metabolite. Moreover, our results indicate that neither induction nor impairment of host glycolysis inhibits intracellular replication of L. pneumophila, which is consistent with a view of L. pneumophila as a metabolic generalist.
Collapse
|
45
|
Mota SGR, Mercaldi GF, Pereira JGC, Oliveira PSL, Rodriguez A, Cordeiro AT. First Nonphosphorylated Inhibitors of Phosphoglucose Isomerase Identified by Chemical Library Screening. SLAS DISCOVERY 2018; 23:1051-1059. [PMID: 29995453 DOI: 10.1177/2472555218787468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Human African trypanosomiasis, Chagas disease, and leishmaniasis are human infections caused by kinetoplastid parasites of the genera Trypanosoma and Leishmania. Besides their severity and global impact, treatments are still challenging. Currently available drugs have important limitations, highlighting the urgent need to develop new drugs. Phosphoglucose isomerase (PGI) is considered a promising target for the development of antiparasitic drugs, as it acts on two essential metabolic pathways, glycolysis and gluconeogenesis. Herein, we describe the identification of new nonphosphorylated inhibitors of Leishmania mexicana PGI ( LmPGI), with the potential for the development of antiparasitic drugs. A fluorescence-based high-throughput screening (HTS) assay was developed by coupling the activities of recombinant LmPGI with glucose-6-phosphate dehydrogenase and diaphorase. This coupled assay was used to screen 42,720 compounds from ChemBridge and TimTec commercial libraries. After confirmatory assays, selected LmPGI inhibitors were tested against homologous Trypanosoma cruzi and humans. The PGI hits are effective against trypanosomatid PGIs, with IC50 values in the micromolar range, and also against the human homologous enzyme. A computational analysis of cavities present on PGI's crystallographic structure suggests a potential binding site for the proposed mixed-type inhibition mechanism.
Collapse
Affiliation(s)
- Sabrina G R Mota
- 1 Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP, Brazil.,2 Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Gustavo F Mercaldi
- 1 Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP, Brazil
| | - José G C Pereira
- 1 Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP, Brazil
| | - Paulo S L Oliveira
- 1 Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP, Brazil
| | - Ana Rodriguez
- 3 New York University School of Medicine, Department of Microbiology, New York, NY, USA
| | - Artur T Cordeiro
- 1 Brazilian Bioscience National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP, Brazil
| |
Collapse
|
46
|
Dumoulin PC, Burleigh BA. Stress-Induced Proliferation and Cell Cycle Plasticity of Intracellular Trypanosoma cruzi Amastigotes. mBio 2018; 9:e00673-18. [PMID: 29991586 PMCID: PMC6050952 DOI: 10.1128/mbio.00673-18] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
The mammalian stages of the parasite Trypanosoma cruzi, the causative agent of Chagas disease, exhibit a wide host species range and extensive within-host tissue distribution. These features, coupled with the ability of the parasites to persist for the lifetime of the host, suggest an inherent capacity to tolerate changing environments. To examine this potential, we studied proliferation and cell cycle dynamics of intracellular T. cruzi amastigotes experiencing transient metabolic perturbation or drug pressure in the context of an infected mammalian host cell. Parasite growth plasticity was evident and characterized by rapid and reversible suppression of amastigote proliferation in response to exogenous nutrient restriction or exposure to metabolic inhibitors that target glucose metabolism or mitochondrial respiration. In most instances, reduced parasite proliferation was accompanied by the accumulation of amastigote populations in the G1 phase of the cell cycle, in a manner that was rapidly and fully reversible upon release from the metabolic block. Acute amastigote cell cycle changes at the G1 stage were similarly observed following exposure to sublethal concentrations of the first-line therapy drug, benznidazole, and yet, unlike the results seen with inhibitors of metabolism, recovery from exposure occurred at rates inversely proportional to the concentration of benznidazole. Our results show that T. cruzi amastigote growth plasticity is an important aspect of parasite adaptation to stress, including drug pressure, and is an important consideration for growth-based drug screening.IMPORTANCE Infection with the intracellular parasite Trypanosoma cruzi can cause debilitating and potentially life-threatening Chagas disease, where long-term parasite persistence is a critical determinant of clinical disease progression. Such tissue-resident T. cruzi amastigotes are refractory to immune-mediated clearance and to drug treatment, suggesting that in addition to exploiting immune avoidance mechanisms, amastigotes can facilitate their survival by adapting flexibly to diverse environmental stressors. We discovered that T. cruzi intracellular amastigotes exhibit growth plasticity as a strategy to adapt to and rebound from environmental stressors, including metabolic blockades, nutrient starvation, and sublethal exposure to the first-line therapy drug benznidazole. These findings have important implications for understanding parasite persistence, informing drug development, and interpreting drug efficacy.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
47
|
The Uptake and Metabolism of Amino Acids, and Their Unique Role in the Biology of Pathogenic Trypanosomatids. Pathogens 2018; 7:pathogens7020036. [PMID: 29614775 PMCID: PMC6027508 DOI: 10.3390/pathogens7020036] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 01/24/2023] Open
Abstract
Trypanosoma brucei, as well as Trypanosoma cruzi and more than 20 species of the genus Leishmania, form a group of flagellated protists that threaten human health. These organisms are transmitted by insects that, together with mammals, are their natural hosts. This implies that during their life cycles each of them faces environments with different physical, chemical, biochemical, and biological characteristics. In this work we review how amino acids are obtained from such environments, how they are metabolized, and how they and some of their intermediate metabolites are used as a survival toolbox to cope with the different conditions in which these parasites should establish the infections in the insects and mammalian hosts.
Collapse
|