1
|
Bonive-Boscan AD, Acosta H, Rojas A. Metabolic changes that allow Plasmodium falciparum artemisinin-resistant parasites to tolerate oxidative stress. FRONTIERS IN PARASITOLOGY 2024; 3:1461641. [PMID: 39817177 PMCID: PMC11731681 DOI: 10.3389/fpara.2024.1461641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 01/18/2025]
Abstract
Artemisinin-based treatments (ACTs) are the first therapy currently used to treat malaria produced by Plasmodium falciparum. However, in recent years, increasing evidence shows that some strains of P. falciparum are less susceptible to ACT in the Southeast Asian region. A data reanalysis of several omics approaches currently available about parasites of P. falciparum that have some degree of resistance to ACT was carried out. The data used were from transcriptomics and metabolomics studies. One mitochondrial carrier of the parasite possibly involved in the mechanisms of tolerance to oxidative stress was modeled and subjected to molecular dockings with citrate and oxoglutarate. An increase in glutathione production was detected, changing the direction of the flux of metabolites in the tricarboxylic acid cycle and boosting the glucose consumed. The models of the mitochondrial carrier, called PfCOCP, show that it may be important in transporting citrate and oxoglutarate from the mitochondrial matrix to the cytosol. If so, it may allow the parasite to tolerate the oxidative stress produced by artemisinin. This in-silico analysis shows that P. falciparum may tolerate artemisinin's oxidative stress through metabolic changes not reported before, showing the need for further experimental research on the many metabolic aspects linked to this phenotype.
Collapse
Affiliation(s)
- Alejandro David Bonive-Boscan
- Centro de Cálculo Científico de la Universidad de Los Andes (CeCalCULA), Universidad de Los Andes (ULA), Mérida, Venezuela
- Max Planck Research Group Evolutionary Cell Biology, Plön, Germany
| | - Héctor Acosta
- Laboratorio de Biología y Bioquímica de Trypanosoma cruzi, Instituto de Biología Molecular y Celular de Rosario, CONICET/UNR, Rosario, Santa Fe, Argentina
| | - Ascanio Rojas
- Centro de Cálculo Científico de la Universidad de Los Andes (CeCalCULA), Universidad de Los Andes (ULA), Mérida, Venezuela
| |
Collapse
|
2
|
Maurizio M, Masid M, Woods K, Caldelari R, Doench JG, Naguleswaran A, Joly D, González-Fernández M, Zemp J, Borteele M, Hatzimanikatis V, Heussler V, Rottenberg S, Olias P. Host cell CRISPR genomics and modelling reveal shared metabolic vulnerabilities in the intracellular development of Plasmodium falciparum and related hemoparasites. Nat Commun 2024; 15:6145. [PMID: 39034325 PMCID: PMC11271486 DOI: 10.1038/s41467-024-50405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Parasitic diseases, particularly malaria (caused by Plasmodium falciparum) and theileriosis (caused by Theileria spp.), profoundly impact global health and the socioeconomic well-being of lower-income countries. Despite recent advances, identifying host metabolic proteins essential for these auxotrophic pathogens remains challenging. Here, we generate a novel metabolic model of human hepatocytes infected with P. falciparum and integrate it with a genome-wide CRISPR knockout screen targeting Theileria-infected cells to pinpoint shared vulnerabilities. We identify key host metabolic enzymes critical for the intracellular survival of both of these lethal hemoparasites. Remarkably, among the metabolic proteins identified by our synergistic approach, we find that host purine and heme biosynthetic enzymes are essential for the intracellular survival of P. falciparum and Theileria, while other host enzymes are only essential under certain metabolic conditions, highlighting P. falciparum's adaptability and ability to scavenge nutrients selectively. Unexpectedly, host porphyrins emerge as being essential for both parasites. The shared vulnerabilities open new avenues for developing more effective therapies against these debilitating diseases, with the potential for broader applicability in combating apicomplexan infections.
Collapse
Affiliation(s)
- Marina Maurizio
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Maria Masid
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne and Lausanne University Teaching Hospital (CHUV), Lausanne, Switzerland
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Kerry Woods
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Denis Joly
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Jonas Zemp
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Mélanie Borteele
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Vassily Hatzimanikatis
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - Philipp Olias
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
3
|
Pandit K, Surolia N, Bhattacharjee S, Karmodiya K. The many paths to artemisinin resistance in Plasmodium falciparum. Trends Parasitol 2023; 39:1060-1073. [PMID: 37833166 DOI: 10.1016/j.pt.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023]
Abstract
Emerging resistance against artemisinin (ART) poses a major challenge in controlling malaria. Parasites with mutations in PfKelch13, the major marker for ART resistance, are known to reduce hemoglobin endocytosis, induce unfolded protein response (UPR), elevate phosphatidylinositol-3-phosphate (PI3P) levels, and stimulate autophagy. Nonetheless, PfKelch13-independent resistance is also reported, indicating extensive complementation by reconfiguration in the parasite metabolome and transcriptome. These findings implicate that there may not be a single 'universal identifier' of ART resistance. This review sheds light on the molecular, transcriptional, and metabolic pathways associated with ART resistance, while also highlighting the interplay between cellular heterogeneity, environmental stress, and ART sensitivity.
Collapse
Affiliation(s)
- Kushankur Pandit
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Namita Surolia
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Souvik Bhattacharjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Krishanpal Karmodiya
- Department of Biology, Indian Institute of Science Education and Research, Pune, India.
| |
Collapse
|
4
|
Brown AC, Warthan MD, Aryal A, Liu S, Guler JL. Nutrient Limitation Mimics Artemisinin Tolerance in Malaria. mBio 2023:e0070523. [PMID: 37097173 DOI: 10.1128/mbio.00705-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Mounting evidence demonstrates that nutritional environment can alter pathogen drug sensitivity. While the rich media used for in vitro culture contains supraphysiological nutrient concentrations, pathogens encounter a relatively restrictive environment in vivo. We assessed the effect of nutrient limitation on the protozoan parasite that causes malaria and demonstrated that short-term growth under physiologically relevant mild nutrient stress (or "metabolic priming") triggers increased tolerance of a potent antimalarial drug. We observed beneficial effects using both short-term survival assays and longer-term proliferation studies, where metabolic priming increases parasite survival to a level previously defined as resistant (>1% survival). We performed these assessments by either decreasing single nutrients that have distinct roles in metabolism or using a media formulation that simulates the human plasma environment. We determined that priming-induced tolerance was restricted to parasites that had newly invaded the host red blood cell, but the effect was not dependent on genetic background. The molecular mechanisms of this intrinsic effect mimic aspects of genetic tolerance, including translational repression and protein export. This finding suggests that regardless of the impact on survival rates, environmental stress could stimulate changes that ultimately directly contribute to drug tolerance. Because metabolic stress is likely to occur more frequently in vivo compared to the stable in vitro environment, priming-induced drug tolerance has ramifications for how in vitro results translate to in vivo studies. Improving our understanding of how pathogens adjust their metabolism to impact survival of current and future drugs is an important avenue of research to slow the evolution of resistance. IMPORTANCE There is a dire need for effective treatments against microbial pathogens. Yet, the continuing emergence of drug resistance necessitates a deeper knowledge of how pathogens respond to treatments. We have long appreciated the contribution of genetic evolution to drug resistance, but transient metabolic changes that arise in response to environmental factors are less recognized. Here, we demonstrate that short-term growth of malaria parasites in a nutrient-limiting environment triggers cellular changes that lead to better survival of drug treatment. We found that these strategies are similar to those employed by drug-tolerant parasites, which suggests that starvation "primes" parasites to survive and potentially evolve resistance. Since the environment of the human host is relatively nutrient restrictive compared to growth conditions in standard laboratory culture, this discovery highlights the important connections among nutrient levels, protective cellular pathways, and resistance evolution.
Collapse
Affiliation(s)
- Audrey C Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Michelle D Warthan
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Anush Aryal
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Shiwei Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Zhu Y, Zhao J, Li J. Genome-scale metabolic modeling in antimicrobial pharmacology. ENGINEERING MICROBIOLOGY 2022; 2:100021. [PMID: 39628842 PMCID: PMC11610950 DOI: 10.1016/j.engmic.2022.100021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/06/2024]
Abstract
The increasing antimicrobial resistance has seriously threatened human health worldwide over the last three decades. This severe medical crisis and the dwindling antibiotic discovery pipeline require the development of novel antimicrobial treatments to combat life-threatening infections caused by multidrug-resistant microbial pathogens. However, the detailed mechanisms of action, resistance, and toxicity of many antimicrobials remain uncertain, significantly hampering the development of novel antimicrobials. Genome-scale metabolic model (GSMM) has been increasingly employed to investigate microbial metabolism. In this review, we discuss the latest progress of GSMM in antimicrobial pharmacology, particularly in elucidating the complex interplays of multiple metabolic pathways involved in antimicrobial activity, resistance, and toxicity. We also highlight the emerging areas of GSMM applications in modeling non-metabolic cellular activities (e.g., gene expression), identification of potential drug targets, and integration with machine learning and pharmacokinetic/pharmacodynamic modeling. Overall, GSMM has significant potential in elucidating the critical role of metabolic changes in antimicrobial pharmacology, providing mechanistic insights that will guide the optimization of dosing regimens for the treatment of antimicrobial-resistant infections.
Collapse
Affiliation(s)
- Yan Zhu
- Infection Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Melbourne, Victoria 3800, Australia
| | - Jinxin Zhao
- Infection Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Melbourne, Victoria 3800, Australia
| | - Jian Li
- Infection Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Melbourne, Victoria 3800, Australia
| |
Collapse
|
6
|
Rawat M, Kanyal A, Choubey D, Deshmukh B, Malhotra R, Mamatharani DV, Rao AG, Karmodiya K. Identification of Co-Existing Mutations and Gene Expression Trends Associated With K13-Mediated Artemisinin Resistance in Plasmodium falciparum. Front Genet 2022; 13:824483. [PMID: 35464842 PMCID: PMC9019836 DOI: 10.3389/fgene.2022.824483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum infects millions and kills thousands of people annually the world over. With the emergence of artemisinin and/or multidrug resistant strains of the pathogen, it has become even more challenging to control and eliminate the disease. Multiomics studies of the parasite have started to provide a glimpse into the confounding genetics and mechanisms of artemisinin resistance and identified mutations in Kelch13 (K13) as a molecular marker of resistance. Over the years, thousands of genomes and transcriptomes of artemisinin-resistant/sensitive isolates have been documented, supplementing the search for new genes/pathways to target artemisinin-resistant isolates. This meta-analysis seeks to recap the genetic landscape and the transcriptional deregulation that demarcate artemisinin resistance in the field. To explore the genetic territory of artemisinin resistance, we use genomic single-nucleotide polymorphism (SNP) datasets from 2,517 isolates from 15 countries from the MalariaGEN Network (The Pf3K project, pilot data release 4, 2015) to dissect the prevalence, geographical distribution, and co-existing patterns of genetic markers associated with/enabling artemisinin resistance. We have identified several mutations which co-exist with the established markers of artemisinin resistance. Interestingly, K13-resistant parasites harbor α-ß hydrolase and putative HECT domain-containing protein genes with the maximum number of SNPs. We have also explored the multiple, publicly available transcriptomic datasets to identify genes from key biological pathways whose consistent deregulation may be contributing to the biology of resistant parasites. Surprisingly, glycolytic and pentose phosphate pathways were consistently downregulated in artemisinin-resistant parasites. Thus, this meta-analysis highlights the genetic and transcriptomic features of resistant parasites to propel further exploratory studies in the community to tackle artemisinin resistance.
Collapse
Affiliation(s)
- Mukul Rawat
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Abhishek Kanyal
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Deepak Choubey
- Life Science Research Unit, Persistent Systems Limited, Pune, India
| | - Bhagyashree Deshmukh
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Rashim Malhotra
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - D V Mamatharani
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Anjani Gopal Rao
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Krishanpal Karmodiya
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| |
Collapse
|
7
|
Carey MA, Medlock GL, Stolarczyk M, Petri WA, Guler JL, Papin JA. Comparative analyses of parasites with a comprehensive database of genome-scale metabolic models. PLoS Comput Biol 2022; 18:e1009870. [PMID: 35196325 PMCID: PMC8901074 DOI: 10.1371/journal.pcbi.1009870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/07/2022] [Accepted: 01/27/2022] [Indexed: 01/01/2023] Open
Abstract
Protozoan parasites cause diverse diseases with large global impacts. Research on the pathogenesis and biology of these organisms is limited by economic and experimental constraints. Accordingly, studies of one parasite are frequently extrapolated to infer knowledge about another parasite, across and within genera. Model in vitro or in vivo systems are frequently used to enhance experimental manipulability, but these systems generally use species related to, yet distinct from, the clinically relevant causal pathogen. Characterization of functional differences among parasite species is confined to post hoc or single target studies, limiting the utility of this extrapolation approach. To address this challenge and to accelerate parasitology research broadly, we present a functional comparative analysis of 192 genomes, representing every high-quality, publicly-available protozoan parasite genome including Plasmodium, Toxoplasma, Cryptosporidium, Entamoeba, Trypanosoma, Leishmania, Giardia, and other species. We generated an automated metabolic network reconstruction pipeline optimized for eukaryotic organisms. These metabolic network reconstructions serve as biochemical knowledgebases for each parasite, enabling qualitative and quantitative comparisons of metabolic behavior across parasites. We identified putative differences in gene essentiality and pathway utilization to facilitate the comparison of experimental findings and discovered that phylogeny is not the sole predictor of metabolic similarity. This knowledgebase represents the largest collection of genome-scale metabolic models for both pathogens and eukaryotes; with this resource, we can predict species-specific functions, contextualize experimental results, and optimize selection of experimental systems for fastidious species.
Collapse
Affiliation(s)
- Maureen A. Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (MAC); (JP)
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Michał Stolarczyk
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Jennifer L. Guler
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jason A. Papin
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (MAC); (JP)
| |
Collapse
|
8
|
Tran T, Rekabdar B, Ekenna C. Deep Learning Methods in Predicting Gene Expression Levels for the Malaria Parasite. Front Genet 2021; 12:721068. [PMID: 34630516 PMCID: PMC8493083 DOI: 10.3389/fgene.2021.721068] [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: 06/28/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
Malaria is a mosquito-borne disease caused by single-celled blood parasites of the genus Plasmodium. The most severe cases of this disease are caused by the Plasmodium species, Falciparum. Once infected, a human host experiences symptoms of recurrent and intermittent fevers occurring over a time-frame of 48 hours, attributed to the synchronized developmental cycle of the parasite during the blood stage. To understand the regulated periodicity of Plasmodium falciparum transcription, this paper forecast and predict the P. falciparum gene transcription during its blood stage life cycle implementing a well-tuned recurrent neural network with gated recurrent units. Additionally, we also employ a spiking neural network to predict the expression levels of the P. falciparum gene. We provide results of this prediction on multiple genes including potential genes that express possible drug target enzymes. Our results show a high level of accuracy in being able to predict and forecast the expression levels of the different genes.
Collapse
Affiliation(s)
- Tuan Tran
- Department of Computer Science, University at Albany, Albany, NY, United States
| | - Banafsheh Rekabdar
- Department of Computer Science, Southern Illinois University, Carbondale, IL, United States
| | - Chinwe Ekenna
- Department of Computer Science, University at Albany, Albany, NY, United States
| |
Collapse
|
9
|
Rodenburg SYA, Seidl MF, de Ridder D, Govers F. Uncovering the Role of Metabolism in Oomycete-Host Interactions Using Genome-Scale Metabolic Models. Front Microbiol 2021; 12:748178. [PMID: 34707596 PMCID: PMC8543037 DOI: 10.3389/fmicb.2021.748178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
Metabolism is the set of biochemical reactions of an organism that enables it to assimilate nutrients from its environment and to generate building blocks for growth and proliferation. It forms a complex network that is intertwined with the many molecular and cellular processes that take place within cells. Systems biology aims to capture the complexity of cells, organisms, or communities by reconstructing models based on information gathered by high-throughput analyses (omics data) and prior knowledge. One type of model is a genome-scale metabolic model (GEM) that allows studying the distributions of metabolic fluxes, i.e., the "mass-flow" through the network of biochemical reactions. GEMs are nowadays widely applied and have been reconstructed for various microbial pathogens, either in a free-living state or in interaction with their hosts, with the aim to gain insight into mechanisms of pathogenicity. In this review, we first introduce the principles of systems biology and GEMs. We then describe how metabolic modeling can contribute to unraveling microbial pathogenesis and host-pathogen interactions, with a specific focus on oomycete plant pathogens and in particular Phytophthora infestans. Subsequently, we review achievements obtained so far and identify and discuss potential pitfalls of current models. Finally, we propose a workflow for reconstructing high-quality GEMs and elaborate on the resources needed to advance a system biology approach aimed at untangling the intimate interactions between plants and pathogens.
Collapse
Affiliation(s)
- Sander Y. A. Rodenburg
- Laboratory of Phytopathology, Wageningen University & Research, Wageningen, Netherlands
- Bioinformatics Group, Wageningen University & Research, Wageningen, Netherlands
| | - Michael F. Seidl
- Laboratory of Phytopathology, Wageningen University & Research, Wageningen, Netherlands
- Theoretical Biology & Bioinformatics group, Department of Biology, Utrecht University, Wageningen, Netherlands
| | - Dick de Ridder
- Bioinformatics Group, Wageningen University & Research, Wageningen, Netherlands
| | - Francine Govers
- Laboratory of Phytopathology, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
10
|
Chiappino-Pepe A, Pandey V, Billker O. Genome reconstructions of metabolism of Plasmodium RBC and liver stages. Curr Opin Microbiol 2021; 63:259-266. [PMID: 34461385 DOI: 10.1016/j.mib.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 11/18/2022]
Abstract
Genome scale metabolic models (GEMs) offer a powerful means of integrating genome and biochemical information on an organism to make testable predictions of metabolic functions at different conditions and to systematically predict essential genes that may be targeted by drugs. This review describes how Plasmodium GEMs have become increasingly more accurate through the integration of omics and experimental genetic data. We also discuss how GEMs contribute to our increasing understanding of how Plasmodium metabolism is reprogrammed between life cycle stages.
Collapse
Affiliation(s)
- Anush Chiappino-Pepe
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vikash Pandey
- Department of Molecular Biology, Umeå University, Umeå, 90187, Sweden; The Laboratory for Molecular Infection Medicine Sweden, Umeå, 90187, Sweden
| | - Oliver Billker
- Department of Molecular Biology, Umeå University, Umeå, 90187, Sweden; The Laboratory for Molecular Infection Medicine Sweden, Umeå, 90187, Sweden
| |
Collapse
|
11
|
Chiappino-Pepe A, Hatzimanikatis V. PhenoMapping: a protocol to map cellular phenotypes to metabolic bottlenecks, identify conditional essentiality, and curate metabolic models. STAR Protoc 2021; 2:100280. [PMID: 33532729 PMCID: PMC7829271 DOI: 10.1016/j.xpro.2020.100280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeted identification of cellular processes responsible for a phenotype is of major importance in guiding efforts in bioengineering and medicine. Genome-scale metabolic models (GEMs) are widely used to integrate various types of omics data and study the cellular physiology under different conditions. Here, we present PhenoMapping, a protocol that uses GEMs, omics, and phenotypic data to map cellular processes and observed phenotypes. PhenoMapping also classifies genes as conditionally and unconditionally essential and guides a comprehensive curation of GEMs. For complete details on the use and execution of this protocol, please refer to Stanway et al. (2019) and Krishnan et al. (2020).
Collapse
Affiliation(s)
- Anush Chiappino-Pepe
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne, Switzerland
| | - Vassily Hatzimanikatis
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne, Switzerland
| |
Collapse
|
12
|
Rawls KD, Dougherty BV, Vinnakota KC, Pannala VR, Wallqvist A, Kolling GL, Papin JA. Predicting changes in renal metabolism after compound exposure with a genome-scale metabolic model. Toxicol Appl Pharmacol 2021; 412:115390. [PMID: 33387578 PMCID: PMC7859602 DOI: 10.1016/j.taap.2020.115390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/02/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
The kidneys are metabolically active organs with importance in several physiological tasks such as the secretion of soluble wastes into the urine and synthesizing glucose and oxidizing fatty acids for energy in fasting (non-fed) conditions. Once damaged, the metabolic capability of the kidneys becomes altered. Here, we define metabolic tasks in a computational modeling framework to capture kidney function in an update to the iRno network reconstruction of rat metabolism using literature-based evidence. To demonstrate the utility of iRno for predicting kidney function, we exposed primary rat renal proximal tubule epithelial cells to four compounds with varying levels of nephrotoxicity (acetaminophen, gentamicin, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six and twenty-four hours, and collected transcriptomics and metabolomics data to measure the metabolic effects of compound exposure. For the transcriptomics data, we observed changes in fatty acid metabolism and amino acid metabolism, as well as changes in existing markers of kidney function such as Clu (clusterin). The iRno metabolic network reconstruction was used to predict alterations in these same pathways after integrating transcriptomics data and was able to distinguish between select compound-specific effects on the proximal tubule epithelial cells. Genome-scale metabolic network reconstructions with coupled omics data can be used to predict changes in metabolism as a step towards identifying novel metabolic biomarkers of kidney function and dysfunction.
Collapse
Affiliation(s)
- Kristopher D Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Bonnie V Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kalyan C Vinnakota
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, MD 20817, USA
| | - Venkat R Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, MD 20817, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA
| | - Glynis L Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
13
|
Mok S, Stokes BH, Gnädig NF, Ross LS, Yeo T, Amaratunga C, Allman E, Solyakov L, Bottrill AR, Tripathi J, Fairhurst RM, Llinás M, Bozdech Z, Tobin AB, Fidock DA. Artemisinin-resistant K13 mutations rewire Plasmodium falciparum's intra-erythrocytic metabolic program to enhance survival. Nat Commun 2021; 12:530. [PMID: 33483501 PMCID: PMC7822823 DOI: 10.1038/s41467-020-20805-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
The emergence and spread of artemisinin resistance, driven by mutations in Plasmodium falciparum K13, has compromised antimalarial efficacy and threatens the global malaria elimination campaign. By applying systems-based quantitative transcriptomics, proteomics, and metabolomics to a panel of isogenic K13 mutant or wild-type P. falciparum lines, we provide evidence that K13 mutations alter multiple aspects of the parasite's intra-erythrocytic developmental program. These changes impact cell-cycle periodicity, the unfolded protein response, protein degradation, vesicular trafficking, and mitochondrial metabolism. K13-mediated artemisinin resistance in the Cambodian Cam3.II line was reversed by atovaquone, a mitochondrial electron transport chain inhibitor. These results suggest that mitochondrial processes including damage sensing and anti-oxidant properties might augment the ability of mutant K13 to protect P. falciparum against artemisinin action by helping these parasites undergo temporary quiescence and accelerated growth recovery post drug elimination.
Collapse
Affiliation(s)
- Sachel Mok
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Barbara H Stokes
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nina F Gnädig
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Leila S Ross
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tomas Yeo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Erik Allman
- Department of Biochemistry & Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, USA
| | - Lev Solyakov
- Protein Nucleic Acid Laboratory, University of Leicester, Leicester, UK
| | - Andrew R Bottrill
- Protein Nucleic Acid Laboratory, University of Leicester, Leicester, UK
| | - Jaishree Tripathi
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Astra Zeneca, Gaithersburg, MD, 20878, USA
| | - Manuel Llinás
- Department of Biochemistry & Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, USA.,Department of Chemistry, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, USA
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Andrew B Tobin
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA. .,Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
14
|
Zou P, Zhang Y, Nshimiyimana JB, Cao Q, Yang Y, Geng H, Xiong L. Reconstruction of a Context-Specific Model Based on Genome-Scale Metabolic Simulation for Identification of Prochloraz Resistance Mechanisms in Penicillium digitatum. Microb Drug Resist 2020; 27:776-785. [PMID: 33180649 DOI: 10.1089/mdr.2020.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Penicillium digitatum is the most destructive postharvest pathogen of citrus fruits, causing substantial economic losses. Prochloraz-resistant strains have emerged due to overuse of imidazole fungicides in agriculture. To study the prochloraz resistance mechanisms at the system level, a genome-scale metabolic model (GEM, iPD1512) of P. digitatum was reconstructed and constrained based on context-specific transcriptome data of the prochloraz-resistant strain, PdF6, from our previous work, a newly sequenced, context-specific transcriptome result of the major facilitator superfamily transporter-encoding gene mfs2 knockout mutant PdF6Δmfs2, and experimentally derived growth rate data. Through the model, iPD1512, the processes of prochloraz resistance in P. digitatum were well simulated. In detail, the growth rates of both wild-type and mutant P. digitatum under different prochloraz concentrations were simulated using constraint-based reconstruction and analysis. The growth rates of the mutant strains (sterol regulatory element-binding protein-encoding gene sreA knockout mutant PdF6ΔsreA and PdF6Δmfs2) were calculated and confirmed to be consistent with the simulation results. Furthermore, correlations between genes and prochloraz resistance were predicted and showed a great difference when compared with correlation results based on p-values from the hypothesis testing used by comparative transcriptomics. To sum up, in contrast to traditional transcriptome analysis, the GEM provides a systemic and dynamic drug resistance mechanism, which might help to detect some key upstream regulatory genes, but with small expression changes, and might provide more efficient targets to control prochloraz-resistant P. digitatum.
Collapse
Affiliation(s)
- Piao Zou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Yunze Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Jean Bosco Nshimiyimana
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Qianwen Cao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Yang Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Hui Geng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Li Xiong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| |
Collapse
|
15
|
Brown AC, Guler JL. From Circulation to Cultivation: Plasmodium In Vivo versus In Vitro. Trends Parasitol 2020; 36:914-926. [DOI: 10.1016/j.pt.2020.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022]
|
16
|
Carey MA, Dräger A, Beber ME, Papin JA, Yurkovich JT. Community standards to facilitate development and address challenges in metabolic modeling. Mol Syst Biol 2020; 16:e9235. [PMID: 32845080 PMCID: PMC8411906 DOI: 10.15252/msb.20199235] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Standardization of data and models facilitates effective communication, especially in computational systems biology. However, both the development and consistent use of standards and resources remain challenging. As a result, the amount, quality, and format of the information contained within systems biology models are not consistent and therefore present challenges for widespread use and communication. Here, we focused on these standards, resources, and challenges in the field of constraint-based metabolic modeling by conducting a community-wide survey. We used this feedback to (i) outline the major challenges that our field faces and to propose solutions and (ii) identify a set of features that defines what a "gold standard" metabolic network reconstruction looks like concerning content, annotation, and simulation capabilities. We anticipate that this community-driven outline will help the long-term development of community-inspired resources as well as produce high-quality, accessible models within our field. More broadly, we hope that these efforts can serve as blueprints for other computational modeling communities to ensure the continued development of both practical, usable standards and reproducible, knowledge-rich models.
Collapse
Affiliation(s)
- Maureen A Carey
- Division of Infectious Diseases and International HealthDepartment of MedicineUniversity of VirginiaCharlottesvilleVAUSA
| | - Andreas Dräger
- Computational Systems Biology of Infection and Antimicrobial‐Resistant PathogensInstitute for Biomedical Informatics (IBMI)University of TübingenTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
- German Center for Infection Research (DZIF), partner site TübingenTübingenGermany
| | - Moritz E Beber
- Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKemitorvetDenmark
| | - Jason A Papin
- Division of Infectious Diseases and International HealthDepartment of MedicineUniversity of VirginiaCharlottesvilleVAUSA
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVAUSA
| | | |
Collapse
|
17
|
Oyelade J, Isewon I, Aromolaran O, Uwoghiren E, Dokunmu T, Rotimi S, Aworunse O, Obembe O, Adebiyi E. Computational Identification of Metabolic Pathways of Plasmodium falciparum using the k-Shortest Path Algorithm. Int J Genomics 2019; 2019:1750291. [PMID: 31662957 PMCID: PMC6791207 DOI: 10.1155/2019/1750291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/28/2018] [Accepted: 07/29/2019] [Indexed: 02/02/2023] Open
Abstract
Plasmodium falciparum, a malaria pathogen, has shown substantial resistance to treatment coupled with poor response to some vaccines thereby requiring urgent, holistic, and broad approach to prevent this endemic disease. Understanding the biology of the malaria parasite has been identified as a vital approach to overcome the threat of malaria. This study is aimed at identifying essential proteins unique to malaria parasites using a reconstructed iPfa genome-scale metabolic model (GEM) of the 3D7 strain of Plasmodium falciparum by filling gaps in the model with nineteen (19) metabolites and twenty-three (23) reactions obtained from the MetaCyc database. Twenty (20) currency metabolites were removed from the network because they have been identified to produce shortcuts that are biologically infeasible. The resulting modified iPfa GEM was a model using the k-shortest path algorithm to identify possible alternative metabolic pathways in glycolysis and pentose phosphate pathways of Plasmodium falciparum. Heuristic function was introduced for the optimal performance of the algorithm. To validate the prediction, the essentiality of the reactions in the reconstructed network was evaluated using betweenness centrality measure, which was applied to every reaction within the pathways considered in this study. Thirty-two (32) essential reactions were predicted among which our method validated fourteen (14) enzymes already predicted in the literature. The enzymatic proteins that catalyze these essential reactions were checked for homology with the host genome, and two (2) showed insignificant similarity, making them possible drug targets. In conclusion, the application of the intelligent search technique to the metabolic network of P. falciparum predicts potential biologically relevant alternative pathways using graph theory-based approach.
Collapse
Affiliation(s)
- Jelili Oyelade
- Department of Computer & Information Sciences, Covenant University, Ota, Nigeria
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
| | - Itunuoluwa Isewon
- Department of Computer & Information Sciences, Covenant University, Ota, Nigeria
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
| | - Olufemi Aromolaran
- Department of Computer & Information Sciences, Covenant University, Ota, Nigeria
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
| | - Efosa Uwoghiren
- Department of Computer & Information Sciences, Covenant University, Ota, Nigeria
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
| | - Titilope Dokunmu
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
- Department of Biochemistry, Covenant University, Ota, Nigeria
| | - Solomon Rotimi
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
- Department of Biochemistry, Covenant University, Ota, Nigeria
| | | | - Olawole Obembe
- Department of Biological Sciences, Covenant University, Ota, Nigeria
| | - Ezekiel Adebiyi
- Department of Computer & Information Sciences, Covenant University, Ota, Nigeria
- Covenant University Bioinformatics Research Cluster (CUBRe), Ota, Nigeria
| |
Collapse
|
18
|
Untaroiu AM, Carey MA, Guler JL, Papin JA. Leveraging the effects of chloroquine on resistant malaria parasites for combination therapies. BMC Bioinformatics 2019; 20:186. [PMID: 30987583 PMCID: PMC6466727 DOI: 10.1186/s12859-019-2756-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 11/10/2022] Open
Abstract
Background Malaria is a major global health problem, with the Plasmodium falciparum protozoan parasite causing the most severe form of the disease. Prevalence of drug-resistant P. falciparum highlights the need to understand the biology of resistance and to identify novel combination therapies that are effective against resistant parasites. Resistance has compromised the therapeutic use of many antimalarial drugs, including chloroquine, and limited our ability to treat malaria across the world. Fortunately, chloroquine resistance comes at a fitness cost to the parasite; this can be leveraged in developing combination therapies or to reinstate use of chloroquine. Results To understand biological changes induced by chloroquine treatment, we compared transcriptomics data from chloroquine-resistant parasites in the presence or absence of the drug. Using both linear models and a genome-scale metabolic network reconstruction of the parasite to interpret the expression data, we identified targetable pathways in resistant parasites. This study identified an increased importance of lipid synthesis, glutathione production/cycling, isoprenoids biosynthesis, and folate metabolism in response to chloroquine. Conclusions We identified potential drug targets for chloroquine combination therapies. Significantly, our analysis predicts that the combination of chloroquine and sulfadoxine-pyrimethamine or fosmidomycin may be more effective against chloroquine-resistant parasites than either drug alone; further studies will explore the use of these drugs as chloroquine resistance blockers. Additional metabolic weaknesses were found in glutathione generation and lipid synthesis during chloroquine treatment. These processes could be targeted with novel inhibitors to reduce parasite growth and reduce the burden of malaria infections. Thus, we identified metabolic weaknesses of chloroquine-resistant parasites and propose targeted chloroquine combination therapies. Electronic supplementary material The online version of this article (10.1186/s12859-019-2756-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana M Untaroiu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.,Present address: Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Maureen A Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,Present address: Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
19
|
Chellapandi P, Prathiviraj R, Prisilla A. Deciphering structure, function and mechanism of Plasmodium IspD homologs from their evolutionary imprints. J Comput Aided Mol Des 2019; 33:419-436. [DOI: 10.1007/s10822-019-00191-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/12/2019] [Indexed: 12/17/2022]
|
20
|
Banerjee D, Raghunathan A. Constraints-based analysis identifies NAD+ recycling through metabolic reprogramming in antibiotic resistant Chromobacterium violaceum. PLoS One 2019; 14:e0210008. [PMID: 30608971 PMCID: PMC6319732 DOI: 10.1371/journal.pone.0210008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Abstract
In the post genomic era, high throughput data augment stoichiometric flux balance models to compute accurate metabolic flux states, growth and energy phenotypes. Investigating altered metabolism in the context of evolved resistant genotypes potentially provide simple strategies to overcome drug resistance and induce susceptibility to existing antibiotics. A genome-scale metabolic model (GSMM) for Chromobacterium violaceum, an opportunistic human pathogen, was reconstructed using legacy data. Experimental constraints were used to represent antibiotic susceptible and resistant populations. Model predictions were validated using growth and respiration data successfully. Differential flux distribution and metabolic reprogramming were identified as a response to antibiotics, chloramphenicol and streptomycin. Streptomycin resistant populations (StrpR) redirected tricarboxylic acid (TCA) cycle flux through the glyoxylate shunt. Chloramphenicol resistant populations (ChlR) resorted to overflow metabolism producing acetate and formate. This switch to fermentative metabolism is potentially through excess reducing equivalents and increased NADH/NAD ratios. Reduced proton gradients and changed Proton Motive Force (PMF) induced by antibiotics were also predicted and verified experimentally using flow cytometry based membrane potential measurements. Pareto analysis of NADH and ATP maintenance showed the decoupling of electron transfer and ATP synthesis in StrpR. Redox homeostasis and NAD+ cycling through rewiring metabolic flux was implicated in re-sensitizing antibiotic resistant C. violaceum. These approaches can be used to probe metabolic vulnerabilities of resistant pathogens. On the verge of a post-antibiotic era, we foresee a critical need for systems level understanding of pathogens and host interaction to extend shelf life of antibiotics and strategize novel therapies.
Collapse
Affiliation(s)
- Deepanwita Banerjee
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| | - Anu Raghunathan
- Chemical Engineering Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| |
Collapse
|
21
|
Dunphy LJ, Yen P, Papin JA. Integrated Experimental and Computational Analyses Reveal Differential Metabolic Functionality in Antibiotic-Resistant Pseudomonas aeruginosa. Cell Syst 2019; 8:3-14.e3. [PMID: 30611675 DOI: 10.1016/j.cels.2018.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/08/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Metabolic adaptations accompanying the development of antibiotic resistance in bacteria remain poorly understood. To study this relationship, we profiled the growth of lab-evolved antibiotic-resistant lineages of the opportunistic pathogen Pseudomonas aeruginosa across 190 unique carbon sources. Our data revealed that the evolution of antibiotic resistance resulted in systems-level changes to growth dynamics and metabolic phenotype. A genome-scale metabolic network reconstruction of P. aeruginosa was paired with whole-genome sequencing data to predict genes contributing to observed changes in metabolism. We experimentally validated computational predictions to identify mutations in resistant P. aeruginosa affecting loss of catabolic function. Finally, we found a shared metabolic phenotype between lab-evolved P. aeruginosa and clinical isolates with similar mutational landscapes. Our results build upon previous knowledge of antibiotic-induced metabolic adaptation and provide a framework for the identification of metabolic limitations in antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Laura J Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Phillip Yen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Medicine, Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA; Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
22
|
Kabra R, Chauhan N, Kumar A, Ingale P, Singh S. Efflux pumps and antimicrobial resistance: Paradoxical components in systems genomics. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 141:15-24. [PMID: 30031023 PMCID: PMC7173168 DOI: 10.1016/j.pbiomolbio.2018.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/10/2018] [Accepted: 07/15/2018] [Indexed: 01/01/2023]
Abstract
Efflux pumps play a major role in the increasing antimicrobial resistance rendering a large number of drugs of no use. Large numbers of pathogens are becoming multidrug resistant due to inadequate dosage and use of the existing antimicrobials. This leads to the need for identifying new efflux pump inhibitors. Design of novel targeted therapies using inherent complexity involved in the biological network modeling has gained increasing importance in recent times. The predictive approaches should be used to determine antimicrobial activities with high pathogen specificity and microbicidal potency. Antimicrobial peptides, which are part of our innate immune system, have the ability to respond to infections and have gained much attention in making resistant strain sensitive to existing drugs. In this review paper, we outline evidences linking host-directed therapy with the efflux pump activity to infectious disease.
Collapse
Affiliation(s)
- Ritika Kabra
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Nutan Chauhan
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Anurag Kumar
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Prajakta Ingale
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| |
Collapse
|
23
|
Zhu Y, Zhao J, Maifiah MHM, Velkov T, Schreiber F, Li J. Metabolic Responses to Polymyxin Treatment in Acinetobacter baumannii ATCC 19606: Integrating Transcriptomics and Metabolomics with Genome-Scale Metabolic Modeling. mSystems 2019; 4:e00157-18. [PMID: 30746493 PMCID: PMC6365644 DOI: 10.1128/msystems.00157-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/08/2019] [Indexed: 02/04/2023] Open
Abstract
Multidrug-resistant (MDR) Acinetobacter baumannii has emerged as a very problematic pathogen over the past decades, with a high incidence in nosocomial infections. Discovered in the late 1940s but abandoned in the 1970s, polymyxins (i.e., polymyxin B and colistin) have been revived as the last-line therapy against Gram-negative "superbugs," including MDR A. baumannii. Worryingly, resistance to polymyxins in A. baumannii has been increasingly reported, urging the development of novel antimicrobial therapies to rescue this last-line class of antibiotics. In the present study, we integrated genome-scale metabolic modeling with multiomics data to elucidate the mechanisms of cellular responses to colistin treatment in A. baumannii. A genome-scale metabolic model, iATCC19606, was constructed for strain ATCC 19606 based on the literature and genome annotation, containing 897 genes, 1,270 reactions, and 1,180 metabolites. After extensive curation, prediction of growth on 190 carbon sources using iATCC19606 achieved an overall accuracy of 84.3% compared to Biolog experimental results. Prediction of gene essentiality reached a high accuracy of 86.1% and 82.7% compared to two transposon mutant libraries of AB5075 and ATCC 17978, respectively. Further integrative modeling with our correlative transcriptomics and metabolomics data deciphered the complex regulation on metabolic responses to colistin treatment, including (i) upregulated fluxes through gluconeogenesis, the pentose phosphate pathway, and amino acid and nucleotide biosynthesis; (ii) downregulated TCA cycle and peptidoglycan and lipopolysaccharide biogenesis; and (iii) altered fluxes over respiratory chain. Our results elucidated the interplay of multiple metabolic pathways under colistin treatment in A. baumannii and provide key mechanistic insights into optimizing polymyxin combination therapy. IMPORTANCE Combating antimicrobial resistance has been highlighted as a critical global health priority. Due to the drying drug discovery pipeline, polymyxins have been employed as the last-line therapy against Gram-negative "superbugs"; however, the detailed mechanisms of antibacterial killing remain largely unclear, hampering the improvement of polymyxin therapy. Our integrative modeling using the constructed genome-scale metabolic model iATCC19606 and the correlative multiomics data provide the fundamental understanding of the complex metabolic responses to polymyxin treatment in A. baumannii at the systems level. The model iATCC19606 may have a significant potential in antimicrobial systems pharmacology research in A. baumannii.
Collapse
Affiliation(s)
- Yan Zhu
- Infection & Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Jinxin Zhao
- Infection & Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Mohd Hafidz Mahamad Maifiah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Tony Velkov
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Jian Li
- Infection & Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| |
Collapse
|
24
|
Chellapandi P, Prathiviraj R, Prisilla A. Molecular evolution and functional divergence of IspD homologs in malarial parasites. INFECTION GENETICS AND EVOLUTION 2018; 65:340-349. [DOI: 10.1016/j.meegid.2018.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 01/19/2023]
|
25
|
Dunphy LJ, Papin JA. Biomedical applications of genome-scale metabolic network reconstructions of human pathogens. Curr Opin Biotechnol 2018; 51:70-79. [PMID: 29223465 PMCID: PMC5991985 DOI: 10.1016/j.copbio.2017.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/14/2022]
Abstract
The growing global threat of antibiotic resistant human pathogens has coincided with improved methods for developing and using genome-scale metabolic network reconstructions. Consequently, there has been an increase in the number of high-quality reconstructions of relevant human and zoonotic pathogens. Novel biomedical applications of pathogen reconstructions focus on three key aspects of pathogen behavior: the evolution of antibiotic resistance, virulence factor production, and host-pathogen interactions. New methods using these reconstructions aim to improve understanding of microbe pathogenicity and guide the development of new therapeutic strategies. This review summarizes the latest ways that genome-scale metabolic network reconstructions have been used to study human pathogens and suggests future applications with the potential to mitigate infectious disease.
Collapse
Affiliation(s)
- Laura J Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA; Department of Medicine, Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
26
|
Influential Parameters for the Analysis of Intracellular Parasite Metabolomics. mSphere 2018; 3:3/2/e00097-18. [PMID: 29669882 PMCID: PMC5907652 DOI: 10.1128/msphere.00097-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/26/2018] [Indexed: 11/20/2022] Open
Abstract
Molecular characterization of pathogens such as the malaria parasite can lead to improved biological understanding and novel treatment strategies. However, the distinctive biology of the Plasmodium parasite, including its repetitive genome and the requirement for growth within a host cell, hinders progress toward these goals. Untargeted metabolomics is a promising approach to learn about pathogen biology. By measuring many small molecules in the parasite at once, we gain a better understanding of important pathways that contribute to the parasite’s response to perturbations such as drug treatment. Although increasingly popular, approaches for intracellular parasite metabolomics and subsequent analysis are not well explored. The findings presented in this report emphasize the critical need for improvements in these areas to limit misinterpretation due to host metabolites and to standardize biological interpretation. Such improvements will aid both basic biological investigations and clinical efforts to understand important pathogens. Metabolomics is increasingly popular for the study of pathogens. For the malaria parasite Plasmodium falciparum, both targeted and untargeted metabolomics have improved our understanding of pathogenesis, host-parasite interactions, and antimalarial drug treatment and resistance. However, purification and analysis procedures for performing metabolomics on intracellular pathogens have not been explored. Here, we purified in vitro-grown ring-stage intraerythrocytic P. falciparum parasites for untargeted metabolomics studies; the small size of this developmental stage amplifies the challenges associated with metabolomics studies as the ratio between host and parasite biomass is maximized. Following metabolite identification and data preprocessing, we explored multiple confounding factors that influence data interpretation, including host contamination and normalization approaches (including double-stranded DNA, total protein, and parasite numbers). We conclude that normalization parameters have large effects on differential abundance analysis and recommend the thoughtful selection of these parameters. However, normalization does not remove the contribution from the parasite’s extracellular environment (culture media and host erythrocyte). In fact, we found that extraparasite material is as influential on the metabolome as treatment with a potent antimalarial drug with known metabolic effects (artemisinin). Because of this influence, we could not detect significant changes associated with drug treatment. Instead, we identified metabolites predictive of host and medium contamination that could be used to assess sample purification. Our analysis provides the first quantitative exploration of the effects of these factors on metabolomics data analysis; these findings provide a basis for development of improved experimental and analytical methods for future metabolomics studies of intracellular organisms. IMPORTANCE Molecular characterization of pathogens such as the malaria parasite can lead to improved biological understanding and novel treatment strategies. However, the distinctive biology of the Plasmodium parasite, including its repetitive genome and the requirement for growth within a host cell, hinders progress toward these goals. Untargeted metabolomics is a promising approach to learn about pathogen biology. By measuring many small molecules in the parasite at once, we gain a better understanding of important pathways that contribute to the parasite’s response to perturbations such as drug treatment. Although increasingly popular, approaches for intracellular parasite metabolomics and subsequent analysis are not well explored. The findings presented in this report emphasize the critical need for improvements in these areas to limit misinterpretation due to host metabolites and to standardize biological interpretation. Such improvements will aid both basic biological investigations and clinical efforts to understand important pathogens.
Collapse
|
27
|
Jayaraman V, Suryavanshi A, Kalale P, Kunala J, Balaram H. Biochemical characterization and essentiality of Plasmodium fumarate hydratase. J Biol Chem 2018; 293:5878-5894. [PMID: 29449371 DOI: 10.1074/jbc.m117.816298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 02/07/2018] [Indexed: 12/30/2022] Open
Abstract
Plasmodium falciparum (Pf), the causative agent of malaria, has an iron-sulfur cluster-containing class I fumarate hydratase (FH) that catalyzes the interconversion of fumarate to malate, a well-known reaction in the tricarboxylic acid cycle. In humans, the same reaction is catalyzed by class II FH that has no sequence or structural homology with the class I enzyme from Plasmodium Fumarate is generated in large quantities in the parasite as a by-product of AMP synthesis and is converted to malate by FH and then used in the generation of the key metabolites oxaloacetate, aspartate, and pyruvate. Previous studies have identified the FH reaction as being essential to P. falciparum, but biochemical characterization of PfFH that may provide leads for the development of specific inhibitors is lacking. Here, we report on the kinetic characterization of purified recombinant PfFH, functional complementation of fh deficiency in Escherichia coli, and mitochondrial localization in the parasite. We found that the substrate analog mercaptosuccinic acid is a potent PfFH inhibitor, with a Ki value in the nanomolar range. The fh gene could not be knocked out in Plasmodium berghei when transfectants were introduced into BALB/c mice; however, fh knockout was successful when C57BL/6 mice were used as host, suggesting that the essentiality of the fh gene to the parasite was mouse strain-dependent.
Collapse
Affiliation(s)
- Vijay Jayaraman
- From the Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, Karnataka 560064, India
| | - Arpitha Suryavanshi
- From the Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, Karnataka 560064, India
| | - Pavithra Kalale
- From the Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, Karnataka 560064, India
| | - Jyothirmai Kunala
- From the Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, Karnataka 560064, India
| | - Hemalatha Balaram
- From the Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, Karnataka 560064, India
| |
Collapse
|
28
|
Abdel-Haleem AM, Hefzi H, Mineta K, Gao X, Gojobori T, Palsson BO, Lewis NE, Jamshidi N. Functional interrogation of Plasmodium genus metabolism identifies species- and stage-specific differences in nutrient essentiality and drug targeting. PLoS Comput Biol 2018; 14:e1005895. [PMID: 29300748 PMCID: PMC5771636 DOI: 10.1371/journal.pcbi.1005895] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/17/2018] [Accepted: 11/24/2017] [Indexed: 12/17/2022] Open
Abstract
Several antimalarial drugs exist, but differences between life cycle stages among malaria species pose challenges for developing more effective therapies. To understand the diversity among stages and species, we reconstructed genome-scale metabolic models (GeMMs) of metabolism for five life cycle stages and five species of Plasmodium spanning the blood, transmission, and mosquito stages. The stage-specific models of Plasmodium falciparum uncovered stage-dependent changes in central carbon metabolism and predicted potential targets that could affect several life cycle stages. The species-specific models further highlight differences between experimental animal models and the human-infecting species. Comparisons between human- and rodent-infecting species revealed differences in thiamine (vitamin B1), choline, and pantothenate (vitamin B5) metabolism. Thus, we show that genome-scale analysis of multiple stages and species of Plasmodium can prioritize potential drug targets that could be both anti-malarials and transmission blocking agents, in addition to guiding translation from non-human experimental disease models. Malaria kills nearly one-half million people a year and over 1 billion people are at risk of becoming infected by the parasite. Plasmodial infections are difficult to treat for a myriad of reasons, but the ability of the organism to remain latent in hosts and the complex life cycles greatly contributed to the difficulty in treat malaria. Genome-scale metabolic models (GeMMs) enable hierarchical integration of disparate data types into a framework amenable to computational simulations enabling deeper mechanistic insights from high-throughput data measurements. In this study, GeMMs of multiple Plasmodium species are used to study metabolic similarities and differences across the Plasmodium genus. In silico gene-knock out simulations across species and stages uncovered functional metabolic differences between human- and rodent-infecting species as well as across the parasite’s life-cycle stages. These findings may help identify drug regimens that are more effective in targeting human-infecting species across multiple stages of the organism.
Collapse
Affiliation(s)
- Alyaa M. Abdel-Haleem
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Centre (CBRC), Thuwal, Saudi Arabia
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering (BESE) division, Thuwal, Saudi Arabia
| | - Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States of America
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, La Jolla, CA, United States of America
| | - Katsuhiko Mineta
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Centre (CBRC), Thuwal, Saudi Arabia
| | - Xin Gao
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Centre (CBRC), Thuwal, Saudi Arabia
| | - Takashi Gojobori
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Centre (CBRC), Thuwal, Saudi Arabia
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States of America
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, La Jolla, CA, United States of America
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
| | - Nathan E. Lewis
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, La Jolla, CA, United States of America
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States of America
| | - Neema Jamshidi
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States of America
- Department of Radiological Sciences, University of California, Los Angeles, CA, United States of America
- * E-mail: ,
| |
Collapse
|