Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Negl Trop Dis ; 10(7): e0004820, 2016 07.
Article in English | MEDLINE | ID: mdl-27467505

ABSTRACT

Cryptosporidium is a major cause of severe diarrhea, especially in malnourished children. Using a murine model of C. parvum oocyst challenge that recapitulates clinical features of severe cryptosporidiosis during malnutrition, we interrogated the effect of protein malnutrition (PM) on primary and secondary responses to C. parvum challenge, and tested the differential ability of mucosal priming strategies to overcome the PM-induced susceptibility. We determined that while PM fundamentally alters systemic and mucosal primary immune responses to Cryptosporidium, priming with C. parvum (106 oocysts) provides robust protective immunity against re-challenge despite ongoing PM. C. parvum priming restores mucosal Th1-type effectors (CD3+CD8+CD103+ T-cells) and cytokines (IFNγ, and IL12p40) that otherwise decrease with ongoing PM. Vaccination strategies with Cryptosporidium antigens expressed in the S. Typhi vector 908htr, however, do not enhance Th1-type responses to C. parvum challenge during PM, even though vaccination strongly boosts immunity in challenged fully nourished hosts. Remote non-specific exposures to the attenuated S. Typhi vector alone or the TLR9 agonist CpG ODN-1668 can partially attenuate C. parvum severity during PM, but neither as effectively as viable C. parvum priming. We conclude that although PM interferes with basal and vaccine-boosted immune responses to C. parvum, sustained reductions in disease severity are possible through mucosal activators of host defenses, and specifically C. parvum priming can elicit impressively robust Th1-type protective immunity despite ongoing protein malnutrition. These findings add insight into potential correlates of Cryptosporidium immunity and future vaccine strategies in malnourished children.


Subject(s)
Cryptosporidiosis/prevention & control , Cryptosporidium/immunology , Dietary Proteins/administration & dosage , Malnutrition/pathology , Protozoan Vaccines/immunology , Administration, Intranasal , Animals , Female , Mice , Mice, Inbred C57BL , Protozoan Vaccines/administration & dosage
2.
PLoS One ; 9(2): e89562, 2014.
Article in English | MEDLINE | ID: mdl-24586873

ABSTRACT

Apolipoliprotein E (apoE), a critical targeting protein in lipid homeostasis, has been found to have immunoinflammatory effects on murine models of infection and malnutrition. The effects of apoE in undernourished and Cryptosporidium parvum-infected mice have not been investigated. In order to study the role of apoE in a model of C. parvum infection, we used the following C57BL6J mouse genetic strains: APOE-deficient, wild-type controls, and APOE targeted replacement (TR) mice expressing human APOE genes (E3/3; E4/4). Experimental mice were orally infected with 10(7)-unexcysted-C. parvum oocysts between post-natal days 34-35 followed by malnutrition induced with a low-protein diet. Mice were euthanized seven days after C. parvum-challenge to investigate ileal morphology, cytokines, and cationic arginine transporter (CAT-1), arginase 1, Toll-like receptor 9 (TLR9), and inducible nitric oxide synthase (iNOS) expression. In addition, we analyzed stool oocyst shedding by qRT-PCR and serum lipids. APOE4/4-TR mice had better weight gains after infection plus malnutrition compared with APOE3/3-TR and wild-type mice. APOE4/4-TR and APOE knockout mice had lower oocyst shedding, however the latter exhibited with villus blunting and higher ileal pro-inflammatory cytokines and iNOS transcripts. APOE4/4-TR mice had increased ileal CAT-1, arginase-1, and TLR9 transcripts relative to APOE knockout. Although with anti-parasitic effects, APOE deficiency exacerbates intestinal inflammatory responses and mucosal damage in undernourished and C. parvum-infected mice. In addition, the human APOE4 gene was found to be protective against the compounded insult of Cryptosporidium infection plus malnutrition, thus extending our previous findings of the protection against diarrhea in APOE4 children. Altogether our findings suggest that apoE plays a key role in the intestinal restitution and immunoinflammatory responses with Cryptosporidium infection and malnutrition.


Subject(s)
Apolipoproteins E/metabolism , Cryptosporidiosis/metabolism , Animals , Apolipoproteins E/genetics , Cryptosporidiosis/genetics , Diet, Protein-Restricted , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic
3.
Virulence ; 4(7): 624-33, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23958904

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is a major pathogen worldwide, associated with diarrheal disease in both children and adults, suggesting the need for new preventive and therapeutic treatments. We investigated the role of the micronutrient zinc in the pathogenesis of an E. coli strain associated with human disease. A variety of bacterial characteristics-growth in vitro, biofilm formation, adherence to IEC-6 epithelial cells, gene expression of putative EAEC virulence factors as well as EAEC-induced cytokine expression by HCT-8 cells-were quantified. At concentrations (≤ 0.05 mM) that did not alter EAEC growth (strain 042) but that are physiologic in serum, zinc markedly decreased the organism's ability to form biofilm (P<0.001), adhere to IEC-6 epithelial cells (P<0.01), and express putative EAEC virulence factors (aggR, aap, aatA, virK) (P<0.03). After exposure of the organism to zinc, the effect on virulence factor generation was prolonged (> 3 h). Further, EAEC-induced IL-8 mRNA and protein secretion by HCT-8 epithelial cells were significantly reduced by 0.05 mM zinc (P<0.03). Using an in vivo murine model of diet-induced zinc-deficiency, oral zinc supplementation (0.4 µg/mouse daily) administered after EAEC challenge (10 (10) CFU/mouse) significantly abrogated growth shortfalls (by>90%; P<0.01); furthermore, stool shedding was reduced (days 9-11) but tissue burden of organisms in the intestine was unchanged. These findings suggest several potential mechanisms whereby physiological levels of zinc alter pathogenetic events in the bacterium (reducing biofilm formation, adherence to epithelium, virulence factor expression) as well as the bacterium's effect on the epithelium (cytokine response to exposure to EAEC) to alter EAEC pathogenesis in vitro and in vivo. These effects may help explain and extend the benefits of zinc in childhood diarrhea and malnutrition.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Biofilms/drug effects , Cytokines/metabolism , Escherichia coli/drug effects , Gene Expression Regulation, Bacterial/drug effects , Zinc/pharmacology , Adult , Animals , Bacterial Load , Bacterial Shedding , Cell Line , Child , Child, Preschool , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/microbiology , Escherichia coli/genetics , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Humans , Intestines/microbiology , Mice , Micronutrients/pharmacology
4.
PLoS One ; 8(2): e57812, 2013.
Article in English | MEDLINE | ID: mdl-23469071

ABSTRACT

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) is recognized as an emerging cause of persistent diarrhea and enteric disease worldwide. Mucosal immunity towards EAEC infections is incompletely understood due in part to the lack of appropriate animal models. This study presents a new mouse model and investigates the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the modulation of host responses to EAEC in nourished and malnourished mice. METHODS/PRINCIPAL FINDINGS: Wild-type and T cell-specific PPARγ null C57BL/6 mice were fed protein-deficient diets at weaning and challenged with 5×10(9)cfu EAEC strain JM221 to measure colonic gene expression and immune responses to EAEC. Antigen-specific responses to E. coli antigens were measured in nourished and malnourished mice following infection and demonstrated the immunosuppressive effects of malnutrition at the cellular level. At the molecular level, both pharmacological blockade and deletion of PPARγ in T cells resulted in upregulation of TGF-ß, IL-6, IL-17 and anti-microbial peptides, enhanced Th17 responses, fewer colonic lesions, faster clearance of EAEC, and improved recovery. The beneficial effects of PPARγ blockade on weight loss and EAEC clearance were abrogated by neutralizing IL-17 in vivo. CONCLUSIONS: Our studies provide in vivo evidence supporting the beneficial role of mucosal innate and effector T cell responses on EAEC burden and suggest pharmacological blockade of PPARγ as a novel therapeutic intervention for EAEC infection.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/physiology , PPAR gamma/metabolism , Anilides/pharmacology , Animals , Antigens, Bacterial/immunology , Bacterial Load/genetics , Bacterial Load/immunology , Body Weight/drug effects , Cell Proliferation/drug effects , Disease Progression , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli/immunology , Escherichia coli Infections/complications , Escherichia coli Infections/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gene Knockout Techniques , Interleukin-17/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Malnutrition/complications , Mice , Mice, Inbred C57BL , PPAR gamma/antagonists & inhibitors , PPAR gamma/deficiency , PPAR gamma/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Time Factors
5.
J Med Microbiol ; 62(Pt 6): 896-905, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23475903

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is increasingly recognized as a common cause of diarrhoea in healthy, malnourished and immune-deficient adults and children. There is no reproducible non-neonatal animal model for longitudinal studies of disease mechanism or therapy. Using two strains of human-derived EAEC to challenge weaned C57BL/6 mice, we explored an in vivo model of EAEC infection in mice, in which disease was monitored quantitatively as the growth rate, stool shedding and tissue burden of organisms; nutritional status was varied, and a new class of therapeutics was assessed. A single oral challenge of EAEC strain 042 resulted in significant growth shortfalls (5-8 % of body weight in 12 days), persistent shedding of micro-organisms in stools [>10(3.2) c.f.u. (10 mg stool)(-1) for at least 14 days] and intestinal tissue burden [~10(3) c.f.u. (10 mg tissue)(-1) detectable up to 14 days post-challenge]. Moderate malnourishment of mice using a 'regional basic diet' containing 7 % protein and reduced fat and micronutrients heightened all parameters of infection. Nitazoxanide in subMIC doses, administered for 3 days at the time of EAEC challenge, lessened growth shortfalls (by >10 % of body weight), stool shedding [by 2-3 logs (10 mg stool)(-1)] and tissue burden of organisms (by >75 % in the jejunum and colon). Thus, weaned C57BL/6 mice challenged with EAEC is a convenient, readily inducible model of EAEC infection with three highly quantifiable outcomes in which disease severity is dependent on the nutritional status of the host, and which is modifiable in the presence of inhibitors of pyruvate ferredoxin oxidoreductase such as nitazoxanide.


Subject(s)
Disease Models, Animal , Escherichia coli Infections/physiopathology , Escherichia coli/pathogenicity , Malnutrition/complications , Thiazoles/therapeutic use , Weaning , Animals , Biofilms/growth & development , Colon/microbiology , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli Infections/complications , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Feces/microbiology , Humans , Male , Mice , Mice, Inbred C57BL , Nitro Compounds , Virulence , Virulence Factors
6.
Vaccine ; 31(6): 912-8, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23246541

ABSTRACT

Cryptosporidium is a protozoan parasite associated with acute and persistent diarrhea that, even in asymptomatic persons, can impair normal growth and potentially cognitive and physical development in young children. The recent availability of the complete gene sequence for Cryptosporidium hominis antigen Cp15 allows examination of innovative vaccine regimens involving intra-nasal antigen priming with live bacterial vectors applicable to human populations. We used a recently described weaned mouse model of cryptosporidiosis, where nourished and malnourished vaccinated mice receive the Cp15 antigen recombinant with cytolysinA on a Salmonella serovar Typhi CVD 908-htr A vector, followed by parenteral exposure to antigen with adjuvant. After challenge with Cryptosporidium oocysts via gavage, parameters of infection and disease (stool shedding of parasites, growth rates) were quantified, and serum/lymphoid tissue harvested to elucidate the Cp15-specific adaptive immune response. In vaccinated nourished mice, the regimen was highly immunogenic, with strong antigen-specific IL-6 and IFN-γ secretion and robust Cp15-specific immunoglobulin titers. In vaccinated malnourished mice, secretion of cytokines, particularly IFN-γ, and antigen-specific humoral immunity were generally undiminished despite protein deprivation and stunted growth. In contrast, after natural (oral) challenge with an identical inoculum of Cryptosporidium oocysts, cytokine and humoral responses to Cp15 were less than one-fourth those in vaccinated mice. Nevertheless, vaccination resulted in only transient reduction in stool shedding of parasites and was not otherwise protective against disease. Overall, immunogenicity for a C. hominis antigen was documented in mice, even in the setting of prolonged malnutrition, using an innovative vaccine regimen involving intra-nasal antigen priming with a live enteric bacterial vector, that has potential applicability to vulnerable human populations irrespective of nutritional status.


Subject(s)
Cytokines/metabolism , Drug Carriers/administration & dosage , Genetic Vectors , Malnutrition/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Salmonella enterica/genetics , Administration, Intranasal , Animals , Antibodies, Helminth/blood , Mice , Mice, Inbred C57BL , Protozoan Proteins/genetics , Protozoan Vaccines/administration & dosage , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
7.
PLoS One ; 7(10): e47525, 2012.
Article in English | MEDLINE | ID: mdl-23071818

ABSTRACT

Clostridium difficile is an anaerobic bacterium that has re-emerged as a facultative pathogen and can cause nosocomial diarrhea, colitis or even death. Peroxisome proliferator-activated receptor (PPAR) γ has been implicated in the prevention of inflammation in autoimmune and infectious diseases; however, its role in the immunoregulatory mechanisms modulating host responses to C. difficile and its toxins remains largely unknown. To characterize the role of PPARγ in C. difficile-associated disease (CDAD), immunity and gut pathology, we used a mouse model of C. difficile infection in wild-type and T cell-specific PPARγ null mice. The loss of PPARγ in T cells increased disease activity and colonic inflammatory lesions following C. difficile infection. Colonic expression of IL-17 was upregulated and IL-10 downregulated in colons of T cell-specific PPARγ null mice. Also, both the loss of PPARγ in T cells and C. difficile infection favored Th17 responses in spleen and colonic lamina propria of mice with CDAD. MicroRNA (miRNA)-sequencing analysis and RT-PCR validation indicated that miR-146b was significantly overexpressed and nuclear receptor co-activator 4 (NCOA4) suppressed in colons of C. difficile-infected mice. We next developed a computational model that predicts the upregulation of miR-146b, downregulation of the PPARγ co-activator NCOA4, and PPARγ, leading to upregulation of IL-17. Oral treatment of C. difficile-infected mice with the PPARγ agonist pioglitazone ameliorated colitis and suppressed pro-inflammatory gene expression. In conclusion, our data indicates that miRNA-146b and PPARγ activation may be implicated in the regulation of Th17 responses and colitis in C. difficile-infected mice.


Subject(s)
Clostridioides difficile/immunology , Colon/pathology , Enterocolitis, Pseudomembranous/immunology , Gene Expression Regulation/immunology , MicroRNAs/immunology , Models, Immunological , PPAR gamma/immunology , Analysis of Variance , Animals , Base Sequence , Colon/immunology , DNA Primers/genetics , Enterocolitis, Pseudomembranous/pathology , High-Throughput Nucleotide Sequencing , Histological Techniques , Immunophenotyping , Interleukin-10/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , PPAR gamma/agonists , Pioglitazone , Real-Time Polymerase Chain Reaction , Th17 Cells/immunology , Thiazolidinediones/pharmacology
8.
J Infect Dis ; 205(9): 1464-71, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22454464

ABSTRACT

BACKGROUND: Although several animal models of cryptosporidiosis have been reported, most involve genetically or pharmacologically immune-suppressed hosts. METHODS: We report challenge with excysted (in vitro and in vivo) and unexcysted (in vivo) Cryptosporidium parvum oocysts in human colonic adenocarcinoma (HCT-8) cells and weaned nourished and malnourished C57BL/6 mice, following outcomes of growth rate, stool shedding, and tissue burden. We tested treatment with an oligodeoxynucleotide containing unmethylated CpG motif (CpG-ODN) and alanyl-glutamine in vivo and in vitro. RESULTS: C. parvum-challenged mice showed prolonged weight loss (>10% over 4 days), robust stool shedding (>3 logs/d over 7 days), and epithelial infection in the ileum, cecum, and colon. Of 2 potential therapeutic compounds evaluated in the model, CpG-ODN reduced body weight loss (to <6% on days 3-7 after challenge), reduced shedding of organisms (by 25% on days 1 and 3 after challenge), and decreased the burden of parasites in the ileum. Alanyl-glutamine showed similar benefits. In vitro findings suggested that effects on the epithelial component of the mucosa probably likely responsible for beneficial effects seen in vivo. CONCLUSIONS: Weaned mice provide a convenient and reproducible model of cryptosporidial disease, including its vicious cycle with body weight loss and heavier infection with malnutrition, and this model may be useful in exploring innovative therapeutic solutions for this challenging infectious disease.


Subject(s)
Cryptosporidiosis/therapy , Cryptosporidium parvum/pathogenicity , Malnutrition/parasitology , Malnutrition/therapy , Animals , Cell Line, Tumor , Colon/drug effects , Colon/parasitology , Colon/pathology , Cryptosporidiosis/complications , Cryptosporidiosis/pathology , Cryptosporidium parvum/isolation & purification , DNA, Protozoan/genetics , DNA, Protozoan/isolation & purification , Dipeptides/therapeutic use , Feces/parasitology , Female , Humans , Ileum/drug effects , Ileum/parasitology , Ileum/pathology , Malnutrition/complications , Malnutrition/pathology , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/therapeutic use
9.
J Parasitol ; 97(6): 1113-20, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21711105

ABSTRACT

Cryptosporidiosis is a leading cause of persistent diarrhea in children in impoverished and developing countries and has both a short- and long-term impact on the growth and development of affected children. An animal model of cryptosporidial infection that mirrors closely the complex interaction between nutritional status and infection in children, particularly in vulnerable settings such as post-weaning and malnourishment, is needed to permit exploration of the pathogenic mechanisms involved. Weaned C57BL/6 mice received a protein-deficient (2%) diet for 3-12 days, then were infected with 5 × 10(7) excysted C. parvum oocyts, and followed for rate of growth, parasite stool shedding, and intestinal invasion/morphometry. Mice had about 20% reduction in weight gain over 12 days of malnutrition and an additional 20% weight loss after C. parvum challenge. Further, a significantly higher fecal C. parvum shedding was detected in malnourished infected mice compared to the nourished infected mice. Also, higher oocyst counts were found in ileum and colon tissue samples from malnourished infected mice, as well as a significant reduction in the villous height-crypt depth ratio in the ileum. Tissue Th1 cytokine concentrations in the ileum were significantly diminished by malnutrition and infection. mRNA for toll-like receptors 2 and 4 were diminished in malnourished infected mice. Treatment with nitazoxanide did not prevent weight loss or parasite stool shedding. These findings indicate that, in the weaned animal, malnutrition intensifies cryptosporidial infection, while cryptosporidial infection further impairs normal growth. Depressed TLR2 and 4 signaling and Th1 cytokine response may be important in the mechanisms underlying the vicious cycle of malnutrition and enteric infection.


Subject(s)
Cryptosporidiosis/complications , Cryptosporidium parvum/pathogenicity , Cytokines/metabolism , Intestinal Mucosa/pathology , Malnutrition/complications , Toll-Like Receptors/metabolism , Animals , Antiparasitic Agents/therapeutic use , Cryptosporidiosis/drug therapy , Cryptosporidiosis/immunology , Cryptosporidiosis/physiopathology , Cryptosporidium parvum/genetics , Cryptosporidium parvum/immunology , DNA, Protozoan/analysis , Disease Models, Animal , Feces/parasitology , Female , Ileum/parasitology , Ileum/pathology , Intestinal Mucosa/parasitology , Malnutrition/immunology , Malnutrition/physiopathology , Mice , Mice, Inbred C57BL , Nitro Compounds , RNA, Messenger/metabolism , Thiazoles/therapeutic use , Toll-Like Receptors/genetics , Weaning , Weight Gain
10.
Am J Trop Med Hyg ; 83(4): 936-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20889896

ABSTRACT

Diarrhea is a major public health problem that affects the development of children. Anthropometric data were collected from 274 children with (N = 170) and without (N = 104) diarrhea. Stool specimens were analyzed by conventional culture, polymerase chain reaction for enteroaggregative Escherichia coli (EAEC), Shigella, Cryptosporidium, Entamoeba, and Giardia species, and by enzyme-linked immunosorbent assay for fecal lactoferrin levels. About 50% of the study population was mildly to severely malnourished. Fecal lactoferrin levels were higher in children with diarrhea (P = 0.019). Children who had EAEC infection, with or without diarrhea, had high mean lactoferrin levels regardless of nutritional status. The EAEC and Cryptosporidium were associated with diarrhea (P = 0.048 and 0.011, respectively), and malnourished children who had diarrhea were often co-infected with both Cryptosporidium and EAEC. In conclusion, the use of DNA-biomarkers revealed that EAEC and Cryptosporidium were common intestinal pathogens in Accra, and that elevated lactoferrin was associated with diarrhea in this group of children.


Subject(s)
Child Nutrition Disorders/complications , Diarrhea/complications , Diarrhea/epidemiology , Gastroenteritis/complications , Case-Control Studies , Child Nutrition Disorders/epidemiology , Child, Preschool , Cross-Sectional Studies , Diarrhea/etiology , Dysentery, Bacillary/complications , Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/microbiology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/complications , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Feces/chemistry , Feces/microbiology , Feces/parasitology , Female , Ghana/epidemiology , Humans , Infant , Infant, Newborn , Lactoferrin/analysis , Male , Parasitic Diseases/complications , Parasitic Diseases/epidemiology , Parasitic Diseases/parasitology , Polymerase Chain Reaction , Prospective Studies , Virulence
11.
J Infect Dis ; 202(4): 506-14, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20594107

ABSTRACT

BACKGROUND: Enteroaggregative Escherichia coli is emerging as an increasingly recognized cause of persistent, mildly inflammatory diarrhea in the United States, especially among patients with AIDS, as well as among children, for whom it is accompanied by growth shortfalls. METHODS: We describe a novel model of disease induced in neonatal and weaned C57BL/6 mice by pathogenic strains of enteroaggregative E. coli 042 and JM221. RESULTS: Enteroaggregative E. coli caused growth impairment (up to 47%), persistent stool shedding (for >3 weeks), and a substantial tissue burden of organisms (150,000 organisms per milligram of tissue), as well as histopathological changes in the colonic epithelium (days 4 and 6 of infection) using the model. Undernutrition in neonatal mice, as well as in weaned mice, intensified infection by 1-4 logs, as assessed by quantitative polymerase chain reaction for fecal shedding of organisms. Growth impairment was dependent on both microorganism burden and challenge dose. CONCLUSIONS: Both neonatal and weaned mice provide models for a vicious cycle of enteroaggregative E. coli infection that causes growth shortfalls and undernutrition, thus worsening infection. Hence, these neonatal and weaned mice provide the opportunity to dissect mechanisms of this cycle in childhood malnutrition, as well as to define the role played by innate and acquired host defenses in this important infection.


Subject(s)
Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Escherichia coli/pathogenicity , Malnutrition/complications , Malnutrition/microbiology , Animals , Animals, Newborn , Colon/microbiology , Colon/pathology , Colony Count, Microbial , Disease Models, Animal , Escherichia coli Infections/microbiology , Feces/microbiology , Female , Growth Disorders/etiology , Histocytochemistry , Humans , Malnutrition/pathology , Mice , Mice, Inbred C57BL , Microscopy , Weaning
12.
Lab Invest ; 88(11): 1178-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18779782

ABSTRACT

Effects in the liver of fatal intoxication with the binary toxin ricin are unclear. We report a robust neutrophil influx into the liver of C57BL/6 mice after lethal parenteral ricin challenge, occurring in peri-portal and centro-lobular hepatic areas within 2 h, followed by the abrupt disappearance of hepatic macrophages/Kupffer cells. Chemokine profiles determined by microarray, ribonuclease protection assays, northern blotting, and enzyme-linked immunosorbent assays showed rapid (2 h) upregulation and persistence of those for neutrophils (CXCL1/KC, CXCL2/MIP-2) and monocytes (CCL2/MCP-1). Red blood cell pooling (8-12 h), loss of hepatocyte glycogen (8-48 h) associated with progressive hypoglycemia, fibrin deposition (24-48 h), and death (72-96 h) followed. Monoclonal antibody to ricin A chain, administered intravenously, blunted hypoglycemia, and abrogated death. This outcome was observed when anti-ricin antibody was given before toxin exposure as well as when administered approximately 10 h after toxin exposure. Targeting antibody to specific amino-acid sequences on the ricin A chain (HAEL and QXXWXXA) was critical to the therapeutic effect. Re-emergence of liver macrophages/Kupffer cells and replenishment of glycogen in previously depleted hepatocytes preceded full recovery of the host. These data identify critical events for liver injury and healing in ricin intoxication, as well as a new means and specific targets for post-exposure therapeutic intervention.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody Specificity/immunology , Chemical Warfare Agents/poisoning , Ricin/immunology , Ricin/poisoning , Animals , Chemical Warfare Agents/chemistry , Disease Models, Animal , Epitopes/immunology , Glycogen/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Kupffer Cells/physiology , Male , Mice , Oligonucleotide Array Sequence Analysis , Poisoning/drug therapy , Poisoning/pathology , Poisoning/physiopathology , Recovery of Function/physiology , Ricin/chemistry
13.
Am J Pathol ; 170(2): 526-37, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255321

ABSTRACT

Neutrophilia is a characteristic of hemolytic uremic syndrome caused by Shiga toxin (Stx2)-producing Escherichia coli. However, the role of neutrophils in the toxin-induced renal injury occurring in enterohemorrhagic E. coli infection remains undefined. We report the trafficking of neutrophils to the kidney of C57BL/6 mice throughout a 72-hour time course after challenge with purified E. coli Stx2 and lipopolysaccharide (LPS). Increased neutrophils were observed in the renal cortex, particularly within the glomeruli where a more than fourfold increase in neutrophils was noted within 2 hours after challenge. Using microarray analysis, an increased number of transcripts for chemoattractants CXCL1/KC (69-fold at 2 hours) and CXCL2/MIP-2 (29-fold at 2 hours) were detected. Ribonuclease protection assays, Northern blotting, enzyme-linked immunosorbent assay, and immunohistochemistry confirmed microarray results, showing that both chemokines were expressed only on the immediate periglomerular epithelium and that these events coincided with neutrophil invasion of glomeruli. Co-administration of Stx2 with LPS enhanced and prolonged the KC and MIP-2 host response (RNA and protein) induced by LPS alone. Immunoneutralization in vivo of CXCL1/KC and CXCL2/MIP-2 abrogated neutrophil migration into glomeruli by 85%. These data define the molecular basis for neutrophil migration into the kidney after exposure to virulence factors of Shiga toxin-producing E. coli O157:H7.


Subject(s)
Chemokines, CXC/biosynthesis , Chemokines/biosynthesis , Escherichia coli Infections/metabolism , Escherichia coli O157 , Nephritis/metabolism , Neutrophils/metabolism , Animals , Cell Movement/drug effects , Chemokine CXCL1 , Chemokine CXCL2 , Escherichia coli Infections/chemically induced , Escherichia coli Infections/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/microbiology , Kidney Glomerulus/pathology , Lipopolysaccharides/toxicity , Male , Mice , Nephritis/chemically induced , Nephritis/microbiology , Nephritis/pathology , Neutrophil Infiltration , Shiga Toxin 2/toxicity
14.
Cancer ; 98(10): 2184-91, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14601088

ABSTRACT

BACKGROUND: Gastric carcinoma is one of the leading causes of cancer-related death worldwide, but the mechanisms underlying its development and progression still remain largely uncharacterized. As loss of trefoil factor 1 (TFF1) expression leads to neoplastic growth in the antropyloric mucosa of mice, the authors sought to comprehensively study the human TFF1 gene in primary gastric carcinomas. METHODS: The authors studied the human TFF1 gene in primary gastric carcinomas and normal gastric mucosa at the DNA, RNA, and protein levels through DNA sequencing, quantitative real-time reverse transcriptase-polymerase chain reaction, and immunohistochemistry. RESULTS: Strikingly, TFF1 mRNA expression was significantly decreased in all 37 gastric carcinomas studied compared with normal gastric mucosa. Furthermore, six tumor/normal pairs with available histologic samples demonstrated a marked decrease in protein expression in tumor samples. Screening of the entire TFF1 coding region in a panel of 42 human gastric tumors did not reveal any somatic mutations, although a few rare germline sequence variants were identified. CONCLUSIONS: These findings demonstrated a significant decrease in the TFF1 transcript in the majority of human gastric carcinomas along with a corresponding reduction in protein expression, both of which occurred in the absence of gene mutation. Dysregulation of TFF1 expression at the transcript level was a critical event in the development of most gastric carcinomas.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Gene Expression Regulation, Neoplastic , Peptides/analysis , Proteins , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Trefoil Factor-1 , Tumor Suppressor Proteins
15.
Magn Reson Med ; 49(6): 1033-6, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12768581

ABSTRACT

In this work, the use of MRI to stage gastric neoplasia in a mouse model of spontaneous gastric cancer is demonstrated. The methodology involves 1) the use of deuterated water ((2)H(2)O) to distend the stomach, and to provide negative contrast between the stomach and the lumen without inducing susceptibility-based field shifts; 2) GlucaGen to minimize motion artifacts that arise from peristalsis; and 3) Gd-DTPA to enhance contrast between the dysplasia/tumor and the normal wall. Initial images are presented from a Tff1 -/- homozygous knock-out model of gastric cancer and a heterozygous C57BL6/J control mouse. There are clear differences between the two types of animals in the MR appearance of the stomach. The distended stomach of the control mouse has a smooth perimeter and a thin wall, and an absence of nodules. The stomach of the Tff1 -/- mouse demonstrates a thickened wall; a jagged, irregular surface; and protruding nodules that are bright after injection of Gd-DTPA.


Subject(s)
Magnetic Resonance Imaging , Stomach Neoplasms/diagnosis , Stomach/pathology , Animals , Contrast Media , Disease Models, Animal , Gadolinium DTPA , Mice , Mice, Knockout
16.
J Cell Physiol ; 194(2): 176-85, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12494456

ABSTRACT

The biology of prostanoids in the normal human colon is only beginning to be understood. We used in situ and in vitro techniques to define the lineage, number, per cell enzyme content, and epithelial functional effect of prostaglandin-generating cells, identified by the presence of cyclooxygenase 1 (COX 1). Immunohistochemical results were quantitated densitometrically, and cell surface staining in situ was verified by flow cytometry of isolated cells and by Western blotting. Three populations of COX 1(+) mucosal cells were identified, based on their morphology and local distribution in human mucosa; these were in the intra-epithelial, crypt apical, and lamina propria regions, with each containing a similar amount of COX 1 protein on a per cell basis. The most numerous were COX 1(+) mononuclear cells in the lamina propria, identified as CD3(+) T lymphocytes, both in situ and ex vivo. In toto, 21% of lamina propria mononuclear cells were COX 1(+), and over 50% of these cells were CD3(+) T cells. Findings were similar in the colon with mild-moderate inflammation due to ulcerative colitis. Using established surface markers, intra-epithelial and crypt apical COX 1(+) cells were non-lymphoid CD45(+) leukocytes; neither IgA (B-lymphocytes) nor alpha-smooth muscle actin (myelofibroblasts) was co-expressed on these COX 1(+) cells. Examining the effect of a major product of COX 1 in an in vitro system of human colonic epithelial monolayers, prostaglandin E(2) (PGE(2)) in low concentration (10(-6) M) enhanced epithelial barrier function and partially protected epithelia from the barrier-disruptive consequences of a pro-inflammatory cytokine, IFN-gamma. We conclude that the human colon contains three tiers of cell types for local synthesis of prostanoids, distinguishable by their location, morphology, and cell lineage. Further, maintenance of the barrier function of colonic epithelium may be added to other cell functions in mucosa regulated, in part, by prostanoids.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Prostaglandins/metabolism , Cell Line , Colitis/metabolism , Colitis/pathology , Colon/cytology , Cyclooxygenase 1 , Dinoprostone/physiology , Flow Cytometry , Humans , Intestinal Mucosa/cytology , Isoenzymes/metabolism , Membrane Proteins , Permeability , Prostaglandin-Endoperoxide Synthases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...