Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell ; 185(24): 4507-4525.e18, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36356582

ABSTRACT

The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.


Subject(s)
Bone Diseases , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Zebrafish , Tuberculosis/microbiology , Macrophages/microbiology , Bacterial Proteins/genetics
2.
Nature ; 602(7895): 106-111, 2022 02.
Article in English | MEDLINE | ID: mdl-34883497

ABSTRACT

Host genetic factors can confer resistance against malaria1, raising the question of whether this has led to evolutionary adaptation of parasite populations. Here we searched for association between candidate host and parasite genetic variants in 3,346 Gambian and Kenyan children with severe malaria caused by Plasmodium falciparum. We identified a strong association between sickle haemoglobin (HbS) in the host and three regions of the parasite genome, which is not explained by population structure or other covariates, and which is replicated in additional samples. The HbS-associated alleles include nonsynonymous variants in the gene for the acyl-CoA synthetase family member2-4 PfACS8 on chromosome 2, in a second region of chromosome 2, and in a region containing structural variation on chromosome 11. The alleles are in strong linkage disequilibrium and have frequencies that covary with the frequency of HbS across populations, in particular being much more common in Africa than other parts of the world. The estimated protective effect of HbS against severe malaria, as determined by comparison of cases with population controls, varies greatly according to the parasite genotype at these three loci. These findings open up a new avenue of enquiry into the biological and epidemiological significance of the HbS-associated polymorphisms in the parasite genome and the evolutionary forces that have led to their high frequency and strong linkage disequilibrium in African P. falciparum populations.


Subject(s)
Genotype , Hemoglobin, Sickle/genetics , Host Adaptation/genetics , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics , Alleles , Animals , Child , Female , Gambia/epidemiology , Genes, Protozoan/genetics , Humans , Kenya/epidemiology , Linkage Disequilibrium , Malaria, Falciparum/epidemiology , Male , Polymorphism, Genetic
3.
mSphere ; 6(5): e0075521, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34668757

ABSTRACT

Sickle-trait hemoglobin (HbAS) confers nearly complete protection from severe, life-threatening falciparum malaria in African children. Despite this clear protection, the molecular mechanisms by which HbAS confers these protective phenotypes remain incompletely understood. As a forward genetic screen for aberrant parasite transcriptional responses associated with parasite neutralization in HbAS red blood cells (RBCs), we performed comparative transcriptomic analyses of Plasmodium falciparum in normal (HbAA) and HbAS erythrocytes during both in vitro cultivation of reference parasite strains and naturally occurring P. falciparum infections in Malian children with HbAA or HbAS. During in vitro cultivation, parasites matured normally in HbAS RBCs, and the temporal expression was largely unperturbed of the highly ordered transcriptional program that underlies the parasite's maturation throughout the intraerythrocytic development cycle (IDC). However, differential expression analysis identified hundreds of transcripts aberrantly expressed in HbAS, largely occurring late in the IDC. Surprisingly, transcripts encoding members of the Maurer's clefts were overexpressed in HbAS despite impaired parasite protein export in these RBCs, while parasites in HbAS RBCs underexpressed transcripts associated with the endoplasmic reticulum and those encoding serine repeat antigen proteases that promote parasite egress. Analyses of P. falciparum transcriptomes from 32 children with uncomplicated malaria identified stage-specific differential expression: among infections composed of ring-stage parasites, only cyclophilin 19B was underexpressed in children with HbAS, while trophozoite-stage infections identified a range of differentially expressed transcripts, including downregulation in HbAS of several transcripts associated with severe malaria in collateral studies. Collectively, our comparative transcriptomic screen in vitro and in vivo indicates that P. falciparum adapts to HbAS by altering its protein chaperone and folding machinery, oxidative stress response, and protein export machinery. Because HbAS consistently protects from severe P. falciparum, modulation of these responses may offer avenues by which to neutralize P. falciparum parasites. IMPORTANCE Sickle-trait hemoglobin (HbAS) confers nearly complete protection from severe, life-threatening malaria, yet the molecular mechanisms that underlie HbAS protection from severe malaria remain incompletely understood. Here, we used transcriptome sequencing (RNA-seq) to measure the impact of HbAS on the blood-stage transcriptome of Plasmodium falciparum in in vitro time series experiments and in vivo samples from natural infections. Our in vitro time series data reveal that, during its blood stage, P. falciparum's gene expression in HbAS is impacted primarily through alterations in the abundance of gene products as opposed to variations in the timing of gene expression. Collectively, our in vitro and in vivo data indicate that P. falciparum adapts to HbAS by altering its protein chaperone and folding machinery, oxidative stress response, and protein export machinery. Due to the persistent association of HbAS and protection from severe disease, these processes that are modified in HbAS may offer strategies to neutralize P. falciparum.


Subject(s)
Hemoglobin A/genetics , Hemoglobin, Sickle/genetics , Malaria, Falciparum/genetics , Sickle Cell Trait/genetics , Adolescent , Child , Child, Preschool , Female , Hemoglobins/metabolism , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Plasmodium falciparum/physiology , Sickle Cell Trait/blood , Sickle Cell Trait/parasitology , Transcriptional Activation
4.
PLoS Pathog ; 17(6): e1009659, 2021 06.
Article in English | MEDLINE | ID: mdl-34115805

ABSTRACT

Sickle-trait hemoglobin protects against severe Plasmodium falciparum malaria. Severe malaria is governed in part by the expression of the Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) that are encoded by var genes, specifically those variants that bind Endothelial Protein C Receptor (EPCR). In this study, we investigate the effect of sickle-trait on parasite var gene expression and function in vitro and in field-collected parasites. We mapped var gene reads generated from RNA sequencing in parasite cultures in normal and sickle-cell trait blood throughout the asexual lifecycle. We investigated sickle-trait effect on PfEMP1 interactions with host receptors CD36 and EPCR using static adhesion assays and flow cytometry. Var expression in vivo was compared by assembling var domains sequenced from total RNA in parasites infecting Malian children with HbAA and HbAS. Sickle-trait did not alter the abundance or type of var gene transcripts in vitro, nor the abundance of overall transcripts or of var functional domains in vivo. In adhesion assays using recombinant host receptors, sickle-trait reduced adhesion by 73-86% to CD36 and 83% to EPCR. Similarly, sickle-trait reduced the surface expression of EPCR-binding PfEMP1. In conclusion, Sickle-cell trait does not directly affect var gene transcription but does reduce the surface expression and function of PfEMP1. This provides a direct mechanism for protection against severe malaria conferred by sickle-trait hemoglobin. Trial Registration: ClinicalTrials.gov Identifier: NCT02645604.


Subject(s)
Hemoglobin, Sickle/metabolism , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , CD36 Antigens/metabolism , Endothelial Protein C Receptor/metabolism , Erythrocytes/parasitology , Hemoglobin, Sickle/genetics , Humans , Malaria, Falciparum/metabolism , Sickle Cell Trait/genetics , Sickle Cell Trait/metabolism
5.
mBio ; 11(3)2020 06 16.
Article in English | MEDLINE | ID: mdl-32546619

ABSTRACT

The ability for cells to maintain homeostasis in the presence of extracellular stress is essential for their survival. Stress adaptations are especially important for microbial pathogens to respond to rapidly changing conditions, such as those encountered during the transition from the environment to the infected host. Many fungal pathogens have acquired the ability to quickly adapt to changes in extracellular pH to promote their survival in the various microenvironments encountered during a host infection. For example, the fungus-specific Rim/Pal alkaline response pathway has been well characterized in many fungal pathogens, including Cryptococcus neoformans However, alternative mechanisms for sensing and responding to host pH have yet to be extensively studied. Recent observations from a genetic screen suggest that the C. neoformans sterol homeostasis pathway is required for growth at elevated pH. This work explores interactions among mechanisms of membrane homeostasis, alkaline pH tolerance, and Rim pathway activation. We find that the sterol homeostasis pathway is necessary for growth in an alkaline environment and that an elevated pH is sufficient to induce Sre1 activation. This pH-mediated activation of the Sre1 transcription factor is linked to the biosynthesis of ergosterol but is not dependent on Rim pathway signaling, suggesting that these two pathways are responding to alkaline pH independently. Furthermore, we discover that C. neoformans is more susceptible to membrane-targeting antifungals under alkaline conditions, highlighting the impact of microenvironmental pH on the treatment of invasive fungal infections. Together, these findings further connect membrane integrity and composition with the fungal pH response and pathogenesis.IMPORTANCE The work described here further elucidates how microorganisms sense and adapt to changes in their environment to establish infections in the human host. Specifically, we uncover a novel mechanism by which an opportunistic human fungal pathogen, Cryptococcus neoformans, responds to increases in extracellular pH in order to survive and thrive within the relatively alkaline environment of the human lung. This mechanism, which is intimately linked with fungal membrane sterol homeostasis, is independent of the previously well-studied alkaline response Rim pathway. Furthermore, this ergosterol-dependent alkaline pH response is present in Candida albicans, indicating that this mechanism spans diverse fungal species. These results are also relevant for novel antimicrobial drug development as we show that currently used ergosterol-targeting antifungals are more active in alkaline environments.


Subject(s)
Cryptococcus neoformans/pathogenicity , Fungal Proteins/metabolism , Host-Pathogen Interactions , Signal Transduction , Sterols/metabolism , Animals , Antifungal Agents/pharmacology , Cell Line , Cryptococcus neoformans/metabolism , Fungal Proteins/genetics , Homeostasis , Hydrogen-Ion Concentration , Mice , Virulence
6.
Nat Commun ; 10(1): 5615, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819062

ABSTRACT

Novel interventions that leverage the heterogeneity of parasite transmission are needed to achieve malaria elimination. To better understand spatial and temporal dynamics of transmission, we applied amplicon next-generation sequencing of two polymorphic gene regions (csp and ama1) to a cohort identified via reactive case detection in a high-transmission setting in western Kenya. From April 2013 to July 2014, we enrolled 442 symptomatic children with malaria, 442 matched controls, and all household members of both groups. Here, we evaluate genetic similarity between infected individuals using three indices: sharing of parasite haplotypes on binary and proportional scales and the L1 norm. Symptomatic children more commonly share haplotypes with their own household members. Furthermore, we observe robust temporal structuring of parasite genetic similarity and identify the unique molecular signature of an outbreak. These findings of both micro- and macro-scale organization of parasite populations might be harnessed to inform next-generation malaria control measures.


Subject(s)
Malaria/epidemiology , Malaria/transmission , Parasites/physiology , Spatio-Temporal Analysis , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Haplotypes/genetics , Humans , Kenya/epidemiology , Middle Aged , Parasites/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Young Adult
8.
Front Immunol ; 10: 528, 2019.
Article in English | MEDLINE | ID: mdl-30967867

ABSTRACT

Over the past 200 years, tuberculosis (TB) has caused more deaths than any other infectious disease, likely infecting more people than it has at any other time in human history. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, is an obligate human pathogen that has evolved through the millennia to become an archetypal human-adapted pathogen. This review focuses on the evolutionary framework by which Mtb emerged as a specialized human pathogen and applies this perspective to the emergence of specific lineages that drive global TB burden. We consider how evolutionary pressures, including transmission dynamics, host tolerance, and human population patterns, may have shaped the evolution of diverse mycobacterial genomes.


Subject(s)
Biological Evolution , Host-Pathogen Interactions/immunology , Immune Tolerance , Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Humans
9.
Mol Microbiol ; 109(4): 474-493, 2018 08.
Article in English | MEDLINE | ID: mdl-29885030

ABSTRACT

The mechanisms by which micro-organisms sense and internalize extracellular pH signals are not completely understood. One example of a known external pH-sensing process is the fungal-specific Rim/Pal signal transduction pathway. Fungi, such as the opportunistic pathogen Cryptococcus neoformans, use Rim signaling to sense and respond to changes in environmental pH. Mutations in this pathway result in strains that are attenuated for survival at alkaline pH, and often for survival within the host. Here, we used an insertional mutagenesis screen to identify novel genes required for C. neoformans growth at host pH. We discovered altered alkaline pH growth in several strains with specific defects in plasma membrane composition and maintenance of phospholipid assembly. Among these, loss of function of the Cdc50 lipid flippase regulatory subunit affected the temporal dynamics of Rim pathway activation. We defined distinct and overlapping cellular processes regulated by Rim101 and Cdc50 through analysis of the transcriptome in these mutant strains. We further explored how pH-induced membrane changes affect membrane-bound pH-sensing proteins, specifically the C-terminal domain of the Rra1 protein, an upstream Rim pathway activator and pH sensor. These results suggest both broadly applicable and phylum-specific molecular interactions that drive microbial environmental sensing.


Subject(s)
Cell Membrane/metabolism , Cryptococcus neoformans/metabolism , Hydrogen-Ion Concentration , Signal Transduction/physiology , Acetyltransferases/metabolism , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Gene Expression Profiling , Mutagenesis, Insertional , P-type ATPases/genetics
10.
Genome Announc ; 6(7)2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29449380

ABSTRACT

Whole-genome sequencing has resulted in new insights into the phylogeography of Mycobacterium tuberculosis However, only limited genomic data are available from M. tuberculosis strains in Guatemala. Here we report 16 complete genomes of clinical strains belonging to the Euro-American lineage 4, the most common lineage found in Guatemala and Central America.

11.
Immunity ; 45(4): 861-876, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760340

ABSTRACT

Mycobacterium tuberculosis infection in humans triggers formation of granulomas, which are tightly organized immune cell aggregates that are the central structure of tuberculosis. Infected and uninfected macrophages interdigitate, assuming an altered, flattened appearance. Although pathologists have described these changes for over a century, the molecular and cellular programs underlying this transition are unclear. Here, using the zebrafish-Mycobacterium marinum model, we found that mycobacterial granuloma formation is accompanied by macrophage induction of canonical epithelial molecules and structures. We identified fundamental macrophage reprogramming events that parallel E-cadherin-dependent mesenchymal-epithelial transitions. Macrophage-specific disruption of E-cadherin function resulted in disordered granuloma formation, enhanced immune cell access, decreased bacterial burden, and increased host survival, suggesting that the granuloma can also serve a bacteria-protective role. Granuloma macrophages in humans with tuberculosis were similarly transformed. Thus, during mycobacterial infection, granuloma macrophages are broadly reprogrammed by epithelial modules, and this reprogramming alters the trajectory of infection and the associated immune response.


Subject(s)
Epithelium/immunology , Macrophages/immunology , Mycobacterium marinum/immunology , Animals , Cadherins/immunology , Epithelium/microbiology , Granuloma/immunology , Granuloma/microbiology , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , Zebrafish
12.
Tuberculosis (Edinb) ; 95(6): 810-816, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26542222

ABSTRACT

Limited data are available regarding the molecular epidemiology of Mycobacterium tuberculosis (Mtb) strains circulating in Guatemala. Beijing-lineage Mtb strains have gained prevalence worldwide and are associated with increased virulence and drug resistance, but there have been only a few cases reported in Central America. Here we report the first whole genome sequencing of Central American Beijing-lineage strains of Mtb. We find that multiple Beijing-lineage strains, derived from independent founding events, are currently circulating in Guatemala, but overall still represent a relatively small proportion of disease burden. Finally, we identify a specific Beijing-lineage outbreak centered on a poor neighborhood in Guatemala City.


Subject(s)
DNA, Bacterial/genetics , Disease Outbreaks , Genome, Bacterial , Mycobacterium tuberculosis/genetics , Sequence Analysis, DNA/methods , Tuberculosis/epidemiology , Urban Health , China/epidemiology , DNA, Bacterial/isolation & purification , Databases, Genetic , Genotype , Guatemala/epidemiology , Humans , Molecular Epidemiology , Mycobacterium tuberculosis/isolation & purification , Mycobacterium tuberculosis/pathogenicity , Phenotype , Phylogeny , Polymorphism, Single Nucleotide , Poverty , Predictive Value of Tests , Tuberculosis/diagnosis , Tuberculosis/microbiology , Tuberculosis/transmission
13.
Dis Model Mech ; 8(12): 1643-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26449262

ABSTRACT

Visualization of infection and the associated host response has been challenging in adult vertebrates. Owing to their transparency, zebrafish larvae have been used to directly observe infection in vivo; however, such larvae have not yet developed a functional adaptive immune system. Cells involved in adaptive immunity mature later and have therefore been difficult to access optically in intact animals. Thus, the study of many aspects of vertebrate infection requires dissection of adult organs or ex vivo isolation of immune cells. Recently, CLARITY and PACT (passive clarity technique) methodologies have enabled clearing and direct visualization of dissected organs. Here, we show that these techniques can be applied to image host-pathogen interactions directly in whole animals. CLARITY and PACT-based clearing of whole adult zebrafish and Mycobacterium tuberculosis-infected mouse lungs enables imaging of mycobacterial granulomas deep within tissue to a depth of more than 1 mm. Using established transgenic lines, we were able to image normal and pathogenic structures and their surrounding host context at high resolution. We identified the three-dimensional organization of granuloma-associated angiogenesis, an important feature of mycobacterial infection, and characterized the induction of the cytokine tumor necrosis factor (TNF) within the granuloma using an established fluorescent reporter line. We observed heterogeneity in TNF induction within granuloma macrophages, consistent with an evolving view of the tuberculous granuloma as a non-uniform, heterogeneous structure. Broad application of this technique will enable new understanding of host-pathogen interactions in situ.


Subject(s)
Aging/pathology , Imaging, Three-Dimensional/methods , Tuberculosis/diagnosis , Tuberculosis/microbiology , Zebrafish/microbiology , Animals , Blood Vessels/pathology , Disease Models, Animal , Female , Fluorescence , Granuloma/microbiology , Granuloma/pathology , Lung/microbiology , Lung/pathology , Mice, Inbred C57BL , Mycobacterium tuberculosis/physiology , Neovascularization, Physiologic , Tuberculosis/pathology , Tumor Necrosis Factor-alpha/metabolism
14.
Clin Vaccine Immunol ; 22(7): 726-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25924766

ABSTRACT

The well-established safety profile of the tuberculosis vaccine strain, Mycobacterium bovis bacille Calmette-Guérin (BCG), makes it an attractive vehicle for heterologous expression of antigens from clinically relevant pathogens. However, successful generation of recombinant BCG strains possessing consistent insert expression has encountered challenges in stability. Here, we describe a method for the development of large recombinant BCG accession lots which stably express the lentiviral antigens, human immunodeficiency virus (HIV) gp120 and simian immunodeficiency virus (SIV) Gag, using selectable leucine auxotrophic complementation. Successful establishment of vaccine stability stems from stringent quality control criteria which not only screen for highly stable complemented BCG ΔleuCD transformants but also thoroughly characterize postproduction quality. These parameters include consistent production of correctly sized antigen, retention of sequence-pure plasmid DNA, freeze-thaw recovery, enumeration of CFU, and assessment of cellular aggregates. Importantly, these quality assurance procedures were indicative of overall vaccine stability, were predictive for successful antigen expression in subsequent passaging both in vitro and in vivo, and correlated with induction of immune responses in murine models. This study has yielded a quality-controlled BCG ΔleuCD vaccine expressing HIV gp120 that retained stable full-length expression after 10(24)-fold amplification in vitro and following 60 days of growth in mice. A second vaccine lot expressed full-length SIV Gag for >10(68)-fold amplification in vitro and induced potent antigen-specific T cell populations in vaccinated mice. Production of large, well-defined recombinant BCG ΔleuCD lots can allow confidence that vaccine materials for immunogenicity and protection studies are not negatively affected by instability or differences between freshly grown production batches.


Subject(s)
Antigens, Viral/biosynthesis , Drug Carriers , Gene Products, gag/biosynthesis , Genomic Instability , HIV Envelope Protein gp120/biosynthesis , Mycobacterium bovis/genetics , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antigens, Viral/genetics , Gene Products, gag/genetics , Genetic Vectors , HIV Envelope Protein gp120/genetics , Mice, Inbred C57BL , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...