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











Publication year range
1.
Curr Top Microbiol Immunol ; 357: 105-36, 2012.
Article in English | MEDLINE | ID: mdl-21983749

ABSTRACT

The kidneys are the major organs affected in diarrhea-associated hemolytic uremic syndrome (D(+)HUS). The pathophysiology of renal disease in D(+)HUS is largely the result of the interaction between bacterial virulence factors such as Shiga toxin and lipopolysaccharide and host cells in the kidney and in the blood circulation. This chapter describes in detail the current knowledge of how these bacterial toxins may lead to kidney disease and renal failure. The toxin receptors expressed by specific blood and resident renal cell types are also discussed as are the actions of the toxins on these cells.


Subject(s)
Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/etiology , Renal Insufficiency/etiology , Shiga Toxin/metabolism , Virulence Factors/metabolism , Blood Cells/metabolism , Fibrinolysis , Glycolipids/metabolism , Hemolytic-Uremic Syndrome/physiopathology , Humans , Inflammation/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/physiopathology , Lipopolysaccharides/metabolism , Renal Insufficiency/physiopathology , Renal Insufficiency/therapy , Sphingolipids/metabolism , Thrombosis/etiology
2.
Toxins (Basel) ; 2(12): 2769-2794, 2010 12 02.
Article in English | MEDLINE | ID: mdl-21297888

ABSTRACT

Shiga toxin-producing Escherichia coli is a contaminant of food and water that in humans causes a diarrheal prodrome followed by more severe disease of the kidneys and an array of symptoms of the central nervous system. The systemic disease is a complex referred to as diarrhea-associated hemolytic uremic syndrome (D(+)HUS). D(+)HUS is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. This review focuses on the renal aspects of D(+)HUS. Current knowledge of this renal disease is derived from a combination of human samples, animal models of D(+)HUS, and interaction of Shiga toxin with isolated renal cell types. Shiga toxin is a multi-subunit protein complex that binds to a glycosphingolipid receptor, Gb3, on select eukaryotic cell types. Location of Gb3 in the kidney is predictive of the sites of action of Shiga toxin. However, the toxin is cytotoxic to some, but not all cell types that express Gb3. It also can cause apoptosis or generate an inflammatory response in some cells. Together, this myriad of results is responsible for D(+)HUS disease.

3.
Infect Immun ; 77(3): 959-69, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19124603

ABSTRACT

Hemolytic-uremic syndrome (HUS) caused by Shiga toxin-producing Escherichia coli infection is a leading cause of pediatric acute renal failure. Bacterial toxins produced in the gut enter the circulation and cause a systemic toxemia and targeted cell damage. It had been previously shown that injection of Shiga toxin 2 (Stx2) and lipopolysaccharide (LPS) caused signs and symptoms of HUS in mice, but the mechanism leading to renal failure remained uncharacterized. The current study elucidated that murine cells of the glomerular filtration barrier were unresponsive to Stx2 because they lacked the receptor glycosphingolipid globotriaosylceramide (Gb(3)) in vitro and in vivo. In contrast to the analogous human cells, Stx2 did not alter inflammatory kinase activity, cytokine release, or cell viability of the murine glomerular cells. However, murine renal cortical and medullary tubular cells expressed Gb(3) and responded to Stx2 by undergoing apoptosis. Stx2-induced loss of functioning collecting ducts in vivo caused production of increased dilute urine, resulted in dehydration, and contributed to renal failure. Stx2-mediated renal dysfunction was ameliorated by administration of the nonselective caspase inhibitor Q-VD-OPH in vivo. Stx2 therefore targets the murine collecting duct, and this Stx2-induced injury can be blocked by inhibitors of apoptosis in vivo.


Subject(s)
Apoptosis/physiology , Epithelium/pathology , Kidney Tubules, Collecting/pathology , Shiga Toxin 2/metabolism , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Blotting, Western , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelium/immunology , Epithelium/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Kidney Tubules, Collecting/immunology , Kidney Tubules, Collecting/metabolism , Lipopolysaccharides/immunology , Mice , Shiga Toxin 2/immunology , Umbilical Veins
4.
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
5.
J Infect Dis ; 198(9): 1398-406, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18754742

ABSTRACT

Affinity-purified Shiga toxin (Stx) 2 given intraperitoneally to mice caused weight loss and hind-limb paralysis followed by death. Globotriaosylceramide (Gb(3)), the receptor for Stx2, was localized to neurons of the central nervous system (CNS) of normal mice. Gb3 was not found in astrocytes or endothelial cells of the CNS. In human cadaver CNS, we found Gb(3) in neurons and endothelial cells. Mouse Gb(3) localization was confirmed by immunoelectron microscopy. In Stx2-exposed mice, anti-Stx2-gold immunoreaction was positive in neurons. During paralysis, after Stx2 injection, multiple glial nuclei were observed surrounding motoneurons by electron microscopy. Also revealed was a lamellipodia-like process physically inhibiting the synaptic connection of motoneurons. Ca2+ imaging of cerebral astrocytic end-feet in Stx2-treated mouse brains suggested that the toxin increased neurotransmitter release from neurons. In this article, we propose that the neuron is a primary target of Stx2, affecting neuronal function and leading to paralysis.


Subject(s)
Central Nervous System/metabolism , Neurons/metabolism , Shiga Toxin 2/toxicity , Trihexosylceramides/metabolism , Animals , Biological Transport , Calcium/metabolism , Central Nervous System/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Male , Mice , Motor Neurons/ultrastructure , Neuroglia/cytology , Specific Pathogen-Free Organisms , Weight Loss
6.
Histochem Cell Biol ; 130(1): 157-64, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18365234

ABSTRACT

Shiga toxin-producing Escherichia coli causes hemolytic uremic syndrome, a constellation of disorders that includes kidney failure and central nervous system dysfunction. Shiga toxin binds the amphipathic, membrane-bound glycolipid globotriaosylceramide (Gb(3)) and uses it to enter host cells and ultimately cause cell death. Thus, cell types that express Gb(3) in target tissues should be recognized. The objective of this study was to determine whether immunohistologic detection of Gb(3) was affected by the method of tissue preparation. Tissue preparation included variations in fixation (immersion or perfusion) and processing (paraffin or frozen) steps; paraffin processing employed different dehydration solvents (acetone or ethanol). Perfusion-fixation in combination with frozen sections or acetone-dehydrated tissue for paraffin sections resulted in specific recognition of Gb(3) using immunohistochemical or immunofluorescent methods. In the mouse tissues studied, Gb(3) was associated with tubules in the kidney and neurons in the nervous system. On the other hand, Gb(3) localization to endothelial cells was determined to be an artifact generated due to immersion-fixation or tissue dehydration with ethanol. This finding was corroborated by glycolipid profiles from tissue subjected to dehydration; namely Gb(3) was subject to extraction by ethanol more than acetone during tissue dehydration. The results of this study show that tissue preparation is crucial to the persistence and preservation of the glycolipid Gb(3) in mouse tissue. These methods may serve as a basis for determining the localization of other amphipathic glycolipids in tissue.


Subject(s)
Fluorescent Antibody Technique, Direct/methods , Frozen Sections/methods , Kidney Tubules/chemistry , Neurons/chemistry , Paraffin Embedding/methods , Trihexosylceramides/analysis , Animals , Antibodies, Monoclonal/immunology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Trihexosylceramides/immunology , Trihexosylceramides/metabolism
7.
Infect Immun ; 76(3): 1115-21, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18086809

ABSTRACT

Escherichia coli O157:H7 Shiga toxin 2 (Stx2), one of the causative agents of hemolytic-uremic syndrome, is toxic to endothelial cells, including primary cultured human umbilical vein endothelial cells (HUVEC). This sensitivity of cells to Stx2 can be increased with either lipopolysaccharide (LPS) or tumor necrosis factor alpha (TNF-alpha). The goal of the present study was to identify the intracellular signaling pathway(s) by which LPS and TNF-alpha sensitize HUVEC to the cytotoxic effects of Stx2. To identify these pathways, specific pharmacological inhibitors and small interfering RNAs were tested with cell viability endpoints. A time course and dose response experiment for HUVEC exposure to LPS and TNF-alpha showed that a relatively short exposure to either agonist was sufficient to sensitize the cells to Stx2 and that both agonists stimulated intracellular signaling pathways within a short time. Cell viability assays indicated that the p38 mitogen-activated protein kinase (MAPK) inhibitors SB202190 and SB203580 and the general protein synthesis inhibitor cycloheximide inhibited both the LPS and TNF-alpha sensitization of HUVEC to Stx2, while all other inhibitors tested did not inhibit this sensitization. Additionally, SB202190 reduced the cellular globotriaosylceramide content under LPS- and TNF-alpha-induced conditions. In conclusion, our results show that LPS and TNF-alpha induction of Stx2 sensitivity in HUVEC is mediated through a pathway that includes p38 MAPK. These results indicate that inhibition of p38 MAPK in endothelial cells may protect a host from the deleterious effects of Stx2.


Subject(s)
Endothelial Cells/microbiology , Escherichia coli O157/physiology , Lipopolysaccharides/metabolism , Shiga Toxin 2/toxicity , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Survival , Cells, Cultured , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/chemistry , Endothelial Cells/immunology , Enzyme Inhibitors/pharmacology , Gene Silencing , Humans , Imidazoles/pharmacology , Pyridines/pharmacology , Time Factors , Trihexosylceramides/analysis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
8.
Infect Immun ; 75(3): 1229-36, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17220320

ABSTRACT

The macrophage has previously been implicated in contributing to the renal inflammation associated with hemolytic-uremic syndrome (HUS). However, there is currently no in vivo model detailing the contribution of the renal macrophage to the kidney disease associated with HUS. Therefore, renal macrophage recruitment and inhibition of infiltrating renal macrophages were evaluated in an established HUS mouse model. Macrophage recruitment to the kidney was evident by immunohistochemistry 2 h after administration of purified Stx2 and peaked at 48 h postinjection. Mice administered a combination of Stx2 and lipopolysaccharide (LPS) showed increased macrophage recruitment to the kidney compared to mice treated with Stx2 or LPS alone. Monocyte chemoattractants were induced in the kidney, including monocyte chemoattractant protein 1 (MCP-1/CCL2), macrophage inflammatory protein 1alpha (MIP-1alpha/CCL3), and RANTES (CCL5), in a pattern that was coincident with macrophage infiltration as indicated by immunohistochemistry, protein, and RNA analyses. MCP-1 was the most abundant chemokine, MIP-1alpha was the least abundant, and RANTES levels were intermediate. Mice treated with MCP-1, MIP-1alpha, and RANTES neutralizing antibodies had a significant decrease in Stx2 plus LPS-induced macrophage accumulation in the kidney, indicating that these chemokines are required for macrophage recruitment. Furthermore, mice exposed to these three neutralizing antibodies had decreased fibrin deposition in their kidneys, implying that macrophages contribute to the renal damage associated with HUS.


Subject(s)
Cell Movement/immunology , Chemokine CCL2/physiology , Chemokine CCL5/physiology , Hemolytic-Uremic Syndrome/metabolism , Kidney/metabolism , Kidney/pathology , Macrophage Inflammatory Proteins/physiology , Macrophages/metabolism , Animals , Chemokine CCL3 , Chemokine CCL4 , Disease Models, Animal , Hemolytic-Uremic Syndrome/immunology , Kidney/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Shiga Toxin 2/pharmacology
9.
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
10.
J Am Soc Nephrol ; 17(12): 3404-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17082244

ABSTRACT

Hemolytic uremic syndrome (HUS), which is caused by Shiga toxin-producing Escherichia coli infection, is the leading cause of acute renal failure in children. At present, there is no complete small animal model of this disease. This study investigated a mouse model using intraperitoneal co-injection of purified Shiga toxin 2 (Stx2) plus LPS. Through microarray, biochemical, and histologic analysis, it was found to be a valid model of the human disease. Biochemical and microarray analysis of mouse kidneys revealed the Stx2 plus LPS challenge to be distinct from the effects of either agent alone. Microarrays identified differentially expressed genes that were demonstrated previously to play a role in this disease. Blood and serum analysis of these mice showed neutrophilia, thrombocytopenia, red cell hemolysis, and increased serum creatinine and blood urea nitrogen. In addition, histologic analysis and electron microscopy of mouse kidneys demonstrated glomerular fibrin deposition, red cell congestion, microthrombi formation, and glomerular ultrastructural changes. It was established that this C57BL/6 mouse is a complete model of HUS that includes the thrombocytopenia, hemolytic anemia, and renal failure that define the human disease. In addition, a time course of HUS disease progression that will be useful for identification of therapeutic targets and development of new treatments for HUS is described.


Subject(s)
Hemolytic-Uremic Syndrome/etiology , Kidney/pathology , Lipopolysaccharides/toxicity , Shiga Toxin 2/toxicity , Anemia, Hemolytic/etiology , Animals , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Gene Expression Regulation/drug effects , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/pathology , Kidney/physiopathology , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Thrombocytopenia/etiology , Weight Loss
11.
Anesth Analg ; 102(4): 1108-13, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16551907

ABSTRACT

Lidocaine attenuates cell injury induced by ischemic-reperfusion and inflammation, although the protective mechanisms are not understood. We hypothesized that lidocaine and other amide local anesthetics protect against endothelial cell injury through activation of the mitochondrial adenosine triphosphate-sensitive potassium (mitoK(ATP)) channels. We determined the effects of amide local anesthetics (lidocaine, ropivacaine, and bupivacaine), ester local anesthetics (tetracaine and procaine), one amide analog (YWI), and two non-amide local anesthetic analogs (JDA and ICM) on viability of human microvascular endothelial cells after exposure to lipopolysaccharide (LPS) in the absence or presence of the mitoK(ATP) channel antagonist 5-hydroxydecaonate. Flavoprotein fluorescence was used to investigate the effects of local anesthetics on diazoxide-induced activation of mitoK(ATP) channels. Lidocaine, ropivacaine, bupivicaine, YWI, JDA, and ICM attenuated by 60% to 70% the decrease in cell viability caused by LPS. Amide local anesthetics and YWI protection was inhibited by 5-hydroxydecaonate, whereas the protection induced by JDA and ICM was not. Tetracaine and procaine did not protect against LPS-induced injury. The amide local anesthetics and the amide analog (YWI) enhanced diazoxide-induced flavoprotein fluorescence by 5% to 20%, whereas ester local anesthetics decreased diazoxide-induced flavoprotein fluorescence by 5% to 60% and the non-amide local anesthetic analogs had no effect. In conclusion, amide local anesthetics and the amide analog (YWI) attenuate LPS-induced cell injury, in part, through activation of mitoK(ATP) channels. In contrast, tetracaine and procaine had no protective effects and inhibited activation of mitoK(ATP) channels. The non-amide local anesthetic analogs induced protection but through mechanisms independent of mitoK(ATP) channels.


Subject(s)
Anesthetics, Local/pharmacology , Cytoprotection/drug effects , Endothelial Cells/drug effects , Lipopolysaccharides/adverse effects , Mitochondria/drug effects , Potassium Channels/physiology , Adenosine Triphosphate/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytoprotection/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Mitochondria/physiology
12.
Pediatr Nephrol ; 18(2): 92-6, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12579394

ABSTRACT

Post-diarrheal (D+) hemolytic uremic syndrome (HUS) is caused by Shiga-toxin (Stx)-producing Escherichia coli. There is epidemiological, cell culture, and mouse model evidence that Stx2-producing E. coli are more likely to cause HUS than strains that produce only Stx1, but this hypothesis has not been tested in a primate model of HUS. We have developed a baboon model of Stx-mediated HUS that was employed to compare the clinical, cytokine, and histological response to equal amounts of the two Shiga toxins. Animals given IV Stx2 developed progressive thrombocytopenia, hemolytic anemia, and azotemia, and urinary interleukin-6 levels rose significantly. Glomerular thrombotic microangiopathy was found at necropsy. Animals given Stx1 showed no cytokine response and no clinical, laboratory, or histological signs of HUS. Our findings from the primate model corroborate previous epidemiological, cell culture, and mouse model observations, and suggest that enteric infection with Stx2-producing E. coli is more likely to cause HUS than infection with organisms that produce only Stx1.


Subject(s)
Hemolytic-Uremic Syndrome/etiology , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Anemia, Hemolytic/etiology , Animals , Disease Models, Animal , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/urine , Interleukin-6/urine , Papio , Thrombocytopenia/etiology , Uremia/etiology
13.
J Immunol ; 169(8): 4496-503, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370386

ABSTRACT

Amebic colitis is an important worldwide parasitic disease for which there is not a well-established animal model. In this work we show that intracecal inoculation of Entamoeba histolytica trophozoites led to established infection in 60% of C3H mice, while C57BL/6 or BALB/c mice were resistant, including mice genetically deficient for IL-12, IFN-gamma, or inducible NO synthase. Infection was a chronic and nonhealing cecitis that pathologically mirrored human disease. Characterization of the inflammation by gene chip analysis revealed abundant mast cell activity. Parasite-specific Ab and cellular proliferative responses were robust and marked by IL-4 and IL-13 production. Depletion of CD4(+) cells significantly diminished both parasite burden and inflammation and correlated with decreased IL-4 and IL-13 production and loss of mast cell infiltration. This model reveals important immune factors that influence susceptibility to infection and demonstrates for the first time the pathologic contribution of the host immune response in amebiasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Disease Models, Animal , Dysentery, Amebic/immunology , Dysentery, Amebic/parasitology , Animals , Antibodies, Protozoan/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Cecum/immunology , Cecum/parasitology , Cecum/pathology , Cell Division/immunology , Chronic Disease , Cricetinae , Disease Susceptibility/immunology , Dysentery, Amebic/pathology , Dysentery, Amebic/prevention & control , Entamoeba histolytica/growth & development , Entamoeba histolytica/immunology , Female , Inflammation/immunology , Inflammation/parasitology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL