Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Emerg Infect Dis ; 19(3): 431-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23622517

ABSTRACT

Noroviruses (NoVs) are a leading cause of gastroenteritis worldwide. An in vitro model for NoV replication remains elusive, making study of the virus difficult. A previous study, which used a 3-dimensional (3-D) intestinal model derived from INT-407 cells reported NoV replication and extensive cytopathic effects (CPE). Using the same 3-D model, but with highly purified Norwalk virus (NV), we attempted to replicate this study. Our results showed no evidence of NV replication by real-time PCR of viral RNA or by immunocytochemical detection of viral structural and nonstructural proteins. Immunocytochemical analysis of the 3-D cultures also showed no detectable presence of histo-blood group antigens that participate in NV binding and host tropism. To determine the potential cause of CPE observed in the previous study, we exposed 3-D cultures to lipopolysaccharide concentrations consistent with contaminated stool samples and observed morphologic features similar to CPE. We conclude that the 3-D INT-407 model does not support NV replication.


Subject(s)
Epithelial Cells/virology , Gastroenteritis/virology , Intestinal Mucosa/virology , Norovirus/physiology , Virus Replication , Blood Group Antigens/metabolism , Cell Aggregation , Cell Culture Techniques , Cell Line , Epithelial Cells/immunology , Gastroenteritis/immunology , Gastroenteritis/pathology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lipopolysaccharides/pharmacology , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Viral Nonstructural Proteins/metabolism , Viral Structural Proteins/metabolism , Viral Tropism
2.
J Infect Dis ; 207(12): 1857-68, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23493725

ABSTRACT

BACKGROUND: Because Mycoplasma genitalium is a prevalent and emerging cause of sexually transmitted infections, understanding the mechanisms by which M. genitalium elicits mucosal inflammation is an essential component to managing lower and upper reproductive tract disease syndromes in women. METHODS: We used a rotating wall vessel bioreactor system to create 3-dimensional (3-D) epithelial cell aggregates to model and assess endocervical infection by M. genitalium. RESULTS: Attachment of M. genitalium to the host cell's apical surface was observed directly and confirmed using immunoelectron microscopy. Bacterial replication was observed from 0 to 72 hours after inoculation, during which time host cells underwent ultrastructural changes, including reduction of microvilli, and marked increases in secretory vesicle formation. Using genome-wide transcriptional profiling, we identified a host defense and inflammation signature activated by M. genitalium during acute infection (48 hours after inoculation) that included cytokine and chemokine activity and secretion of factors for antimicrobial defense. Multiplex bead-based protein assays confirmed secretion of proinflammatory cytokines, several of which are involved in leukocyte recruitment and hypothesized to enhance susceptibility to human immunodeficiency type 1 infection. CONCLUSIONS: These findings provide insight into key molecules and pathways involved in innate recognition of M. genitalium and the response to acute infection in the human endocervix.


Subject(s)
Cervix Uteri/immunology , Cytokines/metabolism , Mycoplasma Infections/immunology , Mycoplasma genitalium/physiology , Sexually Transmitted Diseases, Bacterial/immunology , Bioreactors , Cells, Cultured , Cervix Uteri/cytology , Cervix Uteri/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Female , Genome-Wide Association Study , Host-Pathogen Interactions , Humans , Immunity, Innate , Microscopy, Immunoelectron , Mycoplasma Infections/microbiology , Mycoplasma genitalium/ultrastructure , Sexually Transmitted Diseases, Bacterial/microbiology
3.
Biol Reprod ; 87(6): 132, 2012 Jun.
Article in English | MEDLINE | ID: mdl-23053434

ABSTRACT

Our understanding of the mechanisms that regulate tissue-specific mucosal defense can be limited by the lack of appropriate human in vitro models. The endocervix lies between the microbe-rich vaginal cavity and the relatively sterile endometrium and is a major portal of entry for Chlamydia trachomatis, Neisseria gonorrhoeae, Mycoplasma genitalium, human immunodeficiency virus (HIV), and herpes simplex virus (HSV) infection in women. The endocervix is lined with a simple epithelium, and these cells produce mucus, which plays a key role in immune defense and reproduction. Here we describe the development of a human three-dimensional endocervical epithelial cell model generated by rotating wall vessel bioreactor technology. The model is composed of cellular aggregates that recapitulate major structural and barrier properties essential for the function and protection of the endocervix, including junctional complexes, microvilli, innate immune receptors, antimicrobial peptides, and mucins, the major structural component of mucus. Using this model, we also report, for the first time, that the membrane-associated mucin genes MUC1, MUC4, and MUC16 are differentially regulated in these aggregates by different bacterial and viral products. Differential induction of antimicrobial peptides was also observed with these products. Together these data define unique and flexible innate endocervical immune signatures that follow exposure to microbial products and that likely play a critical role in the outcome of pathogen challenge at this site.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , CA-125 Antigen/metabolism , Cervix Uteri/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Models, Biological , Mucin-1/metabolism , Mucin-4/metabolism , Antimicrobial Cationic Peptides/chemistry , CA-125 Antigen/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cervix Uteri/cytology , Cervix Uteri/ultrastructure , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Female , Humans , Immunity, Mucosal , Ligands , Membrane Proteins/genetics , Mucin-1/genetics , Mucin-4/genetics , Mucins/genetics , Mucins/metabolism , Mucous Membrane/cytology , Mucous Membrane/metabolism , Mucous Membrane/ultrastructure , Secretory Leukocyte Peptidase Inhibitor/genetics , Secretory Leukocyte Peptidase Inhibitor/metabolism , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , beta-Defensins/genetics , beta-Defensins/metabolism
4.
J Vis Exp ; (62)2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22491366

ABSTRACT

Cells and tissues in the body experience environmental conditions that influence their architecture, intercellular communications, and overall functions. For in vitro cell culture models to accurately mimic the tissue of interest, the growth environment of the culture is a critical aspect to consider. Commonly used conventional cell culture systems propagate epithelial cells on flat two-dimensional (2-D) impermeable surfaces. Although much has been learned from conventional cell culture systems, many findings are not reproducible in human clinical trials or tissue explants, potentially as a result of the lack of a physiologically relevant microenvironment. Here, we describe a culture system that overcomes many of the culture condition boundaries of 2-D cell cultures, by using the innovative rotating wall vessel (RWV) bioreactor technology. We and others have shown that organotypic RWV-derived models can recapitulate structure, function, and authentic human responses to external stimuli similarly to human explant tissues (1-6). The RWV bioreactor is a suspension culture system that allows for the growth of epithelial cells under low physiological fluid shear conditions. The bioreactors come in two different formats, a high-aspect rotating vessel (HARV) or a slow-turning lateral vessel (STLV), in which they differ by their aeration source. Epithelial cells are added to the bioreactor of choice in combination with porous, collagen-coated microcarrier beads (Figure 1A). The cells utilize the beads as a growth scaffold during the constant free fall in the bioreactor (Figure 1B). The microenvironment provided by the bioreactor allows the cells to form three-dimensional (3-D) aggregates displaying in vivo-like characteristics often not observed under standard 2-D culture conditions (Figure 1D). These characteristics include tight junctions, mucus production, apical/basal orientation, in vivo protein localization, and additional epithelial cell-type specific properties. The progression from a monolayer of epithelial cells to a fully differentiated 3-D aggregate varies based on cell type(1, 7-13). Periodic sampling from the bioreactor allows for monitoring of epithelial aggregate formation, cellular differentiation markers and viability (Figure 1D). Once cellular differentiation and aggregate formation is established, the cells are harvested from the bioreactor, and similar assays performed on 2-D cells can be applied to the 3-D aggregates with a few considerations (Figure 1E-G). In this work, we describe detailed steps of how to culture 3-D epithelial cell aggregates in the RWV bioreactor system and a variety of potential assays and analyses that can be executed with the 3-D aggregates. These analyses include, but are not limited to, structural/morphological analysis (confocal, scanning and transmission electron microscopy), cytokine/chemokine secretion and cell signaling (cytometric bead array and Western blot analysis), gene expression analysis (real-time PCR), toxicological/drug analysis and host-pathogen interactions. The utilization of these assays set the foundation for more in-depth and expansive studies such as metabolomics, transcriptomics, proteomics and other array-based applications. Our goal is to present a non-conventional means of culturing human epithelial cells to produce organotypic 3-D models that recapitulate the human in vivo tissue, in a facile and robust system to be used by researchers with diverse scientific interests.


Subject(s)
Bioreactors , Cell Culture Techniques/methods , Epithelial Cells/cytology , Humans
5.
Proc Natl Acad Sci U S A ; 108(4): 1633-8, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21220348

ABSTRACT

Virulence of the intracellular pathogen Listeria monocytogenes (Listeria) requires escape from the phagosome into the host cytosol, where the bacteria replicate. Phagosomal escape is a multistep process characterized by perforation, which is dependent on the pore-forming toxin listeriolysin O (LLO), followed by rupture. The contribution of host factors to Listeria phagosomal escape is incompletely defined. Here we show that the cystic fibrosis transmembrane conductance regulator (CFTR) facilitates Listeria cytosolic entry. CFTR inhibition or mutation suppressed Listeria vacuolar escape in culture, and inhibition of CFTR in wild-type mice before oral inoculation of Listeria markedly decreased systemic infection. We provide evidence that high chloride concentrations may facilitate Listeria vacuolar escape by enhancing LLO oligomerization and lytic activity. We propose that CFTR transiently increases phagosomal chloride concentration after infection, potentiating LLO pore formation and vacuole lysis. Our studies suggest that Listeria exploits mechanisms of cellular ion homeostasis to escape the phagosome and emphasize host ion-channel function as a key parameter of bacterial virulence.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Listeria monocytogenes/physiology , Listeriosis/microbiology , Phagosomes/microbiology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Benzoates/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line , Cells, Cultured , Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cytosol/microbiology , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Host-Pathogen Interactions , Hydrazines/pharmacology , Hydrogen-Ion Concentration , Listeria monocytogenes/metabolism , Listeriosis/genetics , Listeriosis/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Multimerization , Thiazolidines/pharmacology , Vacuoles/microbiology , ortho-Aminobenzoates/pharmacology
6.
Nat Rev Microbiol ; 8(11): 791-801, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20948552

ABSTRACT

Appropriately simulating the three-dimensional (3D) environment in which tissues normally develop and function is crucial for engineering in vitro models that can be used for the meaningful dissection of host-pathogen interactions. This Review highlights how the rotating wall vessel bioreactor has been used to establish 3D hierarchical models that range in complexity from a single cell type to multicellular co-culture models that recapitulate the 3D architecture of tissues observed in vivo. The application of these models to the study of infectious diseases is discussed.


Subject(s)
Bioreactors , Host-Pathogen Interactions/physiology , Models, Biological , Animals , Biomedical Engineering , Cells, Cultured , Communicable Diseases/etiology , Humans , In Vitro Techniques , Rotation
7.
PLoS One ; 5(12): e15750, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21206750

ABSTRACT

The prevailing paradigm of Salmonella enteropathogenesis based on monolayers asserts that Salmonella pathogenicity island-1 Type Three Secretion System (SPI-1 T3SS) is required for bacterial invasion into intestinal epithelium. However, little is known about the role of SPI-1 in mediating gastrointestinal disease in humans. Recently, SPI-1 deficient nontyphoidal Salmonella strains were isolated from infected humans and animals, indicating that SPI-1 is not required to cause enteropathogenesis and demonstrating the need for more in vivo-like models. Here, we utilized a previously characterized 3-D organotypic model of human intestinal epithelium to elucidate the role of all characterized Salmonella enterica T3SSs. Similar to in vivo reports, the Salmonella SPI-1 T3SS was not required to invade 3-D intestinal cells. Additionally, Salmonella strains carrying single (SPI-1 or SPI-2), double (SPI-1/2) and complete T3SS knockout (SPI-1/SPI-2: flhDC) also invaded 3-D intestinal cells to wildtype levels. Invasion of wildtype and TTSS mutants was a Salmonella active process, whereas non-invasive bacterial strains, bacterial size beads, and heat-killed Salmonella did not invade 3-D cells. Wildtype and T3SS mutants did not preferentially target different cell types identified within the 3-D intestinal aggregates, including M-cells/M-like cells, enterocytes, or Paneth cells. Moreover, each T3SS was necessary for substantial intracellular bacterial replication within 3-D cells. Collectively, these results indicate that T3SSs are dispensable for Salmonella invasion into highly differentiated 3-D models of human intestinal epithelial cells, but are required for intracellular bacterial growth, paralleling in vivo infection observations and demonstrating the utility of these models in predicting in vivo-like pathogenic mechanisms.


Subject(s)
Intestinal Mucosa/metabolism , Mutation , Salmonella enterica/genetics , Animals , Cell Line , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Humans , Imaging, Three-Dimensional , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Microscopy, Confocal/methods , Salmonella Infections/microbiology
8.
Cell Microbiol ; 10(11): 2197-207, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18665841

ABSTRACT

Membranes are an integral component of many cellular functions and serve as a barrier to keep pathogenic bacteria from entering the nutrient-rich host cytosol. TANK-binding-kinase-1 (TBK1), a kinase of the IkappaB kinase family, is required for maintaining integrity of pathogen-containing vacuoles (PCV) upon bacterial invasion of host cells. Here we investigate how vacuolar integrity is maintained during bacterial infection, even in the presence of bacterial membrane damaging agents. We found that Aquaporin-1 (AQP1), a water channel that regulates swelling of secretory vesicles, associated with PCV. AQP1 levels were elevated in TBK1-deficient cells, and overexpression of AQP1 in wild-type cells led to PCV destabilization, similar to that observed in tbk1(-/-) cells. Inhibition of physiological levels of AQP1 in multiple cell types also led to increased instability of PCV, demonstrating a need for tightly regulated AQP1 function to maintain vacuole homeostasis during bacterial infection. AQP1-dependent modulation of PCV was triggered by bacterially induced membrane damage and ion flux. These results highlight the contribution of water channels to promoting PCV membrane integrity, and reveal an unexpected role for TBK1 and AQP1 in restricting bacterial pathogens to the vacuolar compartment.


Subject(s)
Aquaporin 1/metabolism , Homeostasis , Protein Serine-Threonine Kinases/metabolism , Vacuoles/microbiology , Animals , Aquaporin 1/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/microbiology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Vacuoles/metabolism
9.
PLoS Pathog ; 3(3): e29, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17335348

ABSTRACT

TANK-binding kinase-1 (TBK1) is an integral component of Type I interferon induction by microbial infection. The importance of TBK1 and Type I interferon in antiviral immunity is well established, but the function of TBK1 in bacterial infection is unclear. Upon infection of murine embryonic fibroblasts with Salmonella enterica serovar Typhimurium (Salmonella), more extensive bacterial proliferation was observed in tbk1(-/-) than tbk1(+/+) cells. TBK1 kinase activity was required for restriction of bacterial infection, but interferon regulatory factor-3 or Type I interferon did not contribute to this TBK1-dependent function. In tbk1(-/-)cells, Salmonella, enteropathogenic Escherichia coli, and Streptococcus pyogenes escaped from vacuoles into the cytosol where increased replication occurred, which suggests that TBK1 regulates the integrity of pathogen-containing vacuoles. Knockdown of tbk1 in macrophages and epithelial cells also resulted in increased bacterial localization in the cytosol, indicating that the role of TBK1 in maintaining vacuolar integrity is relevant in different cell types. Taken together, these data demonstrate a requirement for TBK1 in control of bacterial infection distinct from its established role in antiviral immunity.


Subject(s)
Escherichia coli Infections/physiopathology , Protein Serine-Threonine Kinases/physiology , Salmonella Infections, Animal/physiopathology , Streptococcal Infections/physiopathology , Vacuoles/microbiology , Vacuoles/physiology , Animals , Cells, Cultured , Endocytosis/physiology , Escherichia coli Infections/prevention & control , Fibroblasts/microbiology , Gene Expression Regulation , HeLa Cells , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Salmonella Infections, Animal/prevention & control , Streptococcal Infections/prevention & control , Transfection
10.
Cell Microbiol ; 8(11): 1720-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16939532

ABSTRACT

Mammalian innate immunity stimulates antigen-specific immune responses and acts to control infection prior to the onset of adaptive immunity. Some bacterial pathogens replicate within the host cell and are therefore sheltered from some protective aspects of innate immunity such as complement. Here we focus on mechanisms of innate intracellular resistance encountered by bacterial pathogens and how some bacteria can evade destruction by the innate immune system. Major strategies of intracellular antibacterial defence include pathogen compartmentalization and iron limitation. Compartmentalization of pathogens within the host endocytic pathway is critical for generating high local concentrations of antimicrobial molecules, such as reactive oxygen species, and regulating concentrations of divalent cations that are essential for microbial growth. Cytosolic sensing, autophagy, sequestration of essential nutrients and membrane attack by antimicrobial peptides are also discussed.


Subject(s)
Bacteria/pathogenicity , Cytoplasm/immunology , Immunity, Innate/immunology , Animals , Cytoplasm/metabolism , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...