Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
mBio ; 15(8): e0172024, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38995074

ABSTRACT

Infection with the apicomplexan parasite Cryptosporidium is a leading cause of diarrheal disease. Cryptosporidiosis is of particular importance in infants and shows a strong association with malnutrition, both as a risk factor and as a consequence. Cryptosporidium invades and replicates within the small intestine epithelial cells. This is a highly dynamic tissue that is developmentally stratified along the villus axis. New cells emerge from a stem cell niche in the crypt and differentiate into mature epithelial cells while moving toward the villus tip, where they are ultimately shed. Here, we studied the impact of Cryptosporidium infection on this dynamic architecture. Tracing DNA synthesis in pulse-chase experiments in vivo, we quantified the genesis and migration of epithelial cells along the villus. We found proliferation and epithelial migration to be elevated in response to Cryptosporidium infection. Infection also resulted in significant cell loss documented by imaging and molecular assays. Consistent with these observations, single-cell RNA sequencing of infected intestines showed a gain of young and a loss of mature cells. Interestingly, enhanced epithelial cell loss was not a function of enhanced apoptosis of infected cells. To the contrary, Cryptosporidium-infected cells were less likely to be apoptotic than bystanders, and experiments in tissue culture demonstrated that infection provided enhanced resistance to chemically induced apoptosis to the host but not bystander cells. Overall, this study suggests that Cryptosporidium may modulate cell apoptosis and documents pronounced changes in tissue homeostasis due to parasite infection, which may contribute to its long-term impact on the developmental and nutritional state of children. IMPORTANCE: The intestine must balance its roles in digestion and nutrient absorption with the maintenance of an effective barrier to colonization and breach by numerous potential pathogens. An important component of this balance is its constant turnover, which is modulated by a gain of cells due to proliferation and loss due to death or extrusion. Here, we report that Cryptosporidium infection changes the dynamics of this process increasing both gain and loss of enterocytes speeding up the villus elevator. This leads to a much more immature epithelium and a reduction of the number of those cells typically found toward the villus apex best equipped to take up key nutrients including carbohydrates and lipids. These changes in the cellular architecture and physiology of the small intestine may be linked to the profound association between cryptosporidiosis and malnutrition.


Subject(s)
Cryptosporidiosis , Cryptosporidium , Epithelial Cells , Cryptosporidiosis/parasitology , Animals , Epithelial Cells/parasitology , Cryptosporidium/genetics , Cryptosporidium/physiology , Mice , Intestinal Mucosa/parasitology , Apoptosis , Humans , Cell Proliferation , Cell Movement , Intestine, Small/parasitology
2.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38829369

ABSTRACT

Cryptosporidium is an enteric pathogen and a prominent cause of diarrheal disease worldwide. Control of Cryptosporidium requires CD4+ T cells, but how protective CD4+ T cell responses are generated is poorly understood. Here, Cryptosporidium parasites that express MHCII-restricted model antigens were generated to understand the basis for CD4+ T cell priming and effector function. These studies revealed that parasite-specific CD4+ T cells are primed in the draining mesenteric lymph node but differentiate into Th1 cells in the gut to provide local parasite control. Although type 1 conventional dendritic cells (cDC1s) were dispensable for CD4+ T cell priming, they were required for CD4+ T cell gut homing and were a source of IL-12 at the site of infection that promoted local production of IFN-γ. Thus, cDC1s have distinct roles in shaping CD4+ T cell responses to an enteric infection: first, to promote gut homing from the mesLN, and second, to drive effector responses in the intestine.


Subject(s)
CD4-Positive T-Lymphocytes , Cryptosporidiosis , Cryptosporidium , Dendritic Cells , Mice, Inbred C57BL , Animals , Dendritic Cells/immunology , Dendritic Cells/parasitology , Cryptosporidiosis/immunology , Cryptosporidiosis/parasitology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Mice , Cryptosporidium/immunology , Cryptosporidium/physiology , Intestines/immunology , Intestines/parasitology , Interleukin-12/metabolism , Interleukin-12/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Th1 Cells/immunology , Lymph Nodes/immunology , Lymph Nodes/parasitology
3.
Nature ; 630(8015): 174-180, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811723

ABSTRACT

The parasite Cryptosporidium is a leading agent of diarrhoeal disease in young children, and a cause and consequence of chronic malnutrition1,2. There are no vaccines and only limited treatment options3. The parasite infects enterocytes, in which it engages in asexual and sexual replication4, both of which are essential to continued infection and transmission. However, their molecular mechanisms remain largely unclear5. Here we use single-cell RNA sequencing to reveal the gene expression programme of the entire Cryptosporidium parvum life cycle in culture and in infected animals. Diverging from the prevailing model6, we find support for only three intracellular stages: asexual type-I meronts, male gamonts and female gametes. We reveal a highly organized program for the assembly of components at each stage. Dissecting the underlying regulatory network, we identify the transcription factor Myb-M as the earliest determinant of male fate, in an organism that lacks genetic sex determination. Conditional expression of this factor overrides the developmental program and induces widespread maleness, while conditional deletion ablates male development. Both have a profound impact on the infection. A large set of stage-specific genes now provides the opportunity to understand, engineer and disrupt parasite sex and life cycle progression to advance the development of vaccines and treatments.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Gene Expression Regulation , Life Cycle Stages , Transcription, Genetic , Animals , Female , Humans , Male , Mice , Cryptosporidiosis/parasitology , Cryptosporidium parvum/genetics , Cryptosporidium parvum/growth & development , Gene Regulatory Networks , Life Cycle Stages/genetics , Proto-Oncogene Proteins c-myb/genetics , Sex Determination Processes/genetics , Single-Cell Gene Expression Analysis
4.
PLoS Pathog ; 20(5): e1011820, 2024 May.
Article in English | MEDLINE | ID: mdl-38718306

ABSTRACT

The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.


Subject(s)
Cryptosporidiosis , Interferon-gamma , Intestinal Mucosa , Mice, Knockout , Animals , Interferon-gamma/metabolism , Interferon-gamma/immunology , Cryptosporidiosis/immunology , Cryptosporidiosis/parasitology , Mice , Intestinal Mucosa/parasitology , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Cryptosporidium , Epithelial Cells/parasitology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Enterocytes/parasitology , Enterocytes/metabolism , Enterocytes/immunology , Mice, Inbred C57BL , Interferon gamma Receptor , STAT1 Transcription Factor/metabolism , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Signal Transduction
5.
J Infect Dis ; 230(2): 278-280, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38813921
6.
Mucosal Immunol ; 17(3): 387-401, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508522

ABSTRACT

Cryptosporidium causes debilitating diarrheal disease in patients with primary and acquired defects in T cell function. However, it has been a challenge to understand how this infection generates T cell responses and how they mediate parasite control. Here, Cryptosporidium was engineered to express a parasite effector protein (MEDLE-2) that contains the major histocompatibility complex-I restricted SIINFEKL epitope which is recognized by T cell receptor transgenic OT-I(OVA-TCR-I) clusters of differentiation (CD)8+ T cells. These modified parasites induced expansion of endogenous SIINFEKL-specific and OT-I CD8+ T cells that were a source of interferon-gamma (IFN-γ) that could restrict growth of Cryptosporidium. This T cell response was dependent on the translocation of the effector and similar results were observed with another secreted parasite effector (rhoptry protein 1). Although infection and these translocated effector proteins are restricted to intestinal epithelial cells, type 1 conventional dendritic cells were required to generate CD8+ T cell responses to these model antigens. These data sets highlight Cryptosporidium effectors as potential targets of the immune system and suggest that crosstalk between enterocytes and type 1 conventional dendritic cells is crucial for CD8+ T cell responses to Cryptosporidium.


Subject(s)
CD8-Positive T-Lymphocytes , Cryptosporidiosis , Cryptosporidium , Dendritic Cells , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Animals , Cryptosporidiosis/immunology , Mice , Cryptosporidium/immunology , Interferon-gamma/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/immunology , Antigens, Protozoan/immunology , Humans , Mice, Transgenic , Lymphocyte Activation/immunology , Epitopes, T-Lymphocyte/immunology , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Mice, Knockout
7.
Nat Commun ; 15(1): 379, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38191574

ABSTRACT

In Apicomplexa, rhoptry discharge is essential for invasion and involves an apical vesicle (AV) docking one or two rhoptries to a macromolecular secretory apparatus. Toxoplasma gondii is armed with 10-12 rhoptries and 5-6 microtubule-associated vesicles (MVs) presumably for iterative rhoptry discharge. Here, we have addressed the localization and functional significance of two intraconoidal microtubule (ICMT)-associated proteins instrumental for invasion. Mechanistically, depletion of ICMAP2 leads to a dissociation of the ICMTs, their detachment from the conoid and dispersion of MVs and rhoptries. ICMAP3 exists in two isoforms that contribute to the control of the ICMTs length and the docking of the two rhoptries at the AV, respectively. This study illuminates the central role ICMTs play in scaffolding the discharge of multiple rhoptries. This process is instrumental for virulence in the mouse model of infection and in addition promotes sterile protection against T. gondii via the release of key effectors inducing immunity.


Subject(s)
Toxoplasma , Animals , Mice , Microtubule-Associated Proteins , Cytoskeleton , Microtubules , Biological Transport
8.
Proc Natl Acad Sci U S A ; 121(1): e2313210120, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38147547

ABSTRACT

Parasites and their hosts are engaged in reciprocal coevolution that balances competing mechanisms of virulence, resistance, and evasion. This often leads to host specificity, but genomic reassortment between different strains can enable parasites to jump host barriers and conquer new niches. In the apicomplexan parasite Cryptosporidium, genetic exchange has been hypothesized to play a prominent role in adaptation to humans. The sexual lifecycle of the parasite provides a potential mechanism for such exchange; however, the boundaries of Cryptosporidium sex are currently undefined. To explore this experimentally, we established a model for genetic crosses. Drug resistance was engineered using a mutated phenylalanyl tRNA synthetase gene and marking strains with this and the previously used Neo transgene enabled selection of recombinant progeny. This is highly efficient, and genomic recombination is evident and can be continuously monitored in real time by drug resistance, flow cytometry, and PCR mapping. Using this approach, multiple loci can now be modified with ease. We demonstrate that essential genes can be ablated by crossing a Cre recombinase driver strain with floxed strains. We further find that genetic crosses are also feasible between species. Crossing Cryptosporidium parvum, a parasite of cattle and humans, and Cryptosporidium tyzzeri a mouse parasite resulted in progeny with a recombinant genome derived from both species that continues to vigorously replicate sexually. These experiments have important fundamental and translational implications for the evolution of Cryptosporidium and open the door to reverse- and forward-genetic analysis of parasite biology and host specificity.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Crosses, Genetic , Cryptosporidiosis/parasitology , Cryptosporidium/genetics , Cryptosporidium parvum/genetics , Life Cycle Stages
SELECTION OF CITATIONS
SEARCH DETAIL