Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(8): 3221-3228, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718423

ABSTRACT

The cell cycle machinery controls diverse cellular pathways and is tightly regulated. Misregulation of cell division plays a central role in the pathogenesis of many disease processes. Various microbial pathogens interfere with the cell cycle machinery to promote host cell colonization. Although cell cycle modulation is a common theme among pathogens, the role this interference plays in promoting diseases is unclear. Previously, we demonstrated that the G1 and G2/M phases of the host cell cycle are permissive for Legionella pneumophila replication, whereas S phase provides a toxic environment for bacterial replication. In this study, we show that L. pneumophila avoids host S phase by blocking host DNA synthesis and preventing cell cycle progression into S phase. Cell cycle arrest upon Legionella contact is dependent on the Icm/Dot secretion system. In particular, we found that cell cycle arrest is dependent on the intact enzymatic activity of translocated substrates that inhibits host translation. Moreover, we show that, early in infection, the presence of these translation inhibitors is crucial to induce the degradation of the master regulator cyclin D1. Our results demonstrate that the bacterial effectors that inhibit translation are associated with preventing entry of host cells into a phase associated with restriction of L. pneumophila Furthermore, control of cyclin D1 may be a common strategy used by intracellular pathogens to manipulate the host cell cycle and promote bacterial replication.


Subject(s)
Cell Cycle Checkpoints/genetics , Cyclin D1/genetics , Host-Pathogen Interactions/genetics , Legionella pneumophila/genetics , DNA Replication/genetics , Humans , Immunity, Innate/genetics , Legionella pneumophila/pathogenicity , Legionnaires' Disease/genetics , Legionnaires' Disease/microbiology , Macrophages/metabolism , Translocation, Genetic/genetics
2.
Cell Host Microbe ; 24(2): 208-220.e8, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30092198

ABSTRACT

In enteric viral infections, such as those with rotavirus and norovirus, individual viral particles shed in stool are considered the optimal units of fecal-oral transmission. We reveal that rotaviruses and noroviruses are also shed in stool as viral clusters enclosed within vesicles that deliver a high inoculum to the receiving host. Cultured cells non-lytically release rotaviruses and noroviruses inside extracellular vesicles. In addition, stools of infected hosts contain norovirus and rotavirus within vesicles of exosomal or plasma membrane origin. These vesicles remain intact during fecal-oral transmission and thereby transport multiple viral particles collectively to the next host, enhancing both the MOI and disease severity. Vesicle-cloaked viruses are non-negligible populations in stool and have a disproportionately larger contribution to infectivity than free viruses. Our findings indicate that vesicle-cloaked viruses are highly virulent units of fecal-oral transmission and highlight a need for antivirals targeting vesicles and virus clustering.


Subject(s)
Caliciviridae Infections/transmission , Extracellular Vesicles/virology , Feces/virology , Rotavirus Infections/transmission , Animals , Caliciviridae Infections/virology , Child, Preschool , Disease Transmission, Infectious , Exosomes/virology , Female , Humans , Male , Mice, Inbred BALB C , Norovirus/genetics , Norovirus/pathogenicity , Rotavirus/genetics , Rotavirus/pathogenicity , Rotavirus Infections/virology , Swine , Virus Shedding
3.
mBio ; 8(4)2017 08 22.
Article in English | MEDLINE | ID: mdl-28830950

ABSTRACT

Legionella pneumophila grows within cells ranging from environmental amoebae to human macrophages. In spite of this conserved strategy of pathogenesis, identification of host factors that restrict L. pneumophila intracellular replication has not been extended outside components of the mammalian innate immune response. We performed a double-stranded RNA (dsRNA) screen against more than 50% of the Drosophila melanogaster annotated open reading frames (ORFs) to identify host cell factors that restrict L. pneumophila The majority of analyzed dsRNAs that stimulated L. pneumophila intracellular replication were directed against host proteins involved in protein synthesis or cell cycle control. Consistent with disruption of the cell cycle stimulating intracellular replication, proteins involved in translation initiation also resulted in G1 arrest. Stimulation of replication was dependent on the stage of cell cycle arrest, as dsRNAs causing arrest during S phase had an inhibitory effect on intracellular replication. The inhibitory effects of S phase arrest could be recapitulated in a human cell line, indicating that cell cycle control of L. pneumophila replication is evolutionarily conserved. Synchronized HeLa cell populations in S phase and challenged with L. pneumophila failed to progress through the cell cycle and were depressed for supporting intracellular replication. Poor bacterial replication in S phase was associated with loss of the vacuole membrane barrier, resulting in exposure of bacteria to the cytosol and their eventual degradation. These results are consistent with the model that S phase is inhibitory for L. pneumophila intracellular survival as a consequence of failure to maintain the integrity of the membrane surrounding intracellular bacteria.IMPORTANCELegionella pneumophila has the ability to replicate within human macrophages and amoebal hosts. Here, we report that the host cell cycle influences L. pneumophila intracellular replication. Our data demonstrate that the G1 and G2/M phases of the host cell cycle are permissive for bacterial replication, while S phase is toxic for the bacterium. L. pneumophila replicates poorly within host cells present in S phase. The inability of L. pneumophila to replicate relies on its failure to control the integrity of its vacuole, leading to cytosolic exposure of the bacteria and eventual degradation. The data presented here argue that growth-arrested host cells that are encountered by L. pneumophila in either the environment or within human hosts are ideal targets for intracellular replication because their transit through S phase is blocked.


Subject(s)
Cytosol/microbiology , DNA Replication , Host-Pathogen Interactions , Legionella pneumophila/physiology , Membranes/metabolism , S Phase , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Cycle/physiology , Cytosol/physiology , Drosophila/genetics , HeLa Cells , Humans , Legionella pneumophila/genetics , Macrophages/microbiology , Membranes/pathology , Open Reading Frames , RNA, Double-Stranded , Vacuoles/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...