Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Histochem Cytochem ; 64(12): 753-767, 2016 12.
Article in English | MEDLINE | ID: mdl-27798287

ABSTRACT

The adenomatous polyposis coli (APC) gene is a known tumor suppressor gene, and mice with mutations in Apc (ApcMin/+) spontaneously form multiple intestinal neoplasms. In this model of human colorectal cancer (CRC), it has been reported that CD4+ T-cell-derived interleukin 17 (IL-17) promotes intestinal tumor development, but it is not known if the Apc mutation actually directly alters T-cell function and subsequently tumor immunosurveillance. To investigate the ApcMin/+ mutation on T-cell function, flow cytometric, histochemical, and immunofluorescent studies on both wild-type (Apc+/+) and ApcMin/+ mice were performed. We identified decreased levels of interferon gamma (IFN-γ+)IL-17+ double-positive CD4+ cells in the mesenteric lymph nodes and Peyer's patches of ApcMin/+ mice. In addition, altered levels of CD8+ cells, and changes in CD8+ production of IFN-γ and granzyme B were observed. These T-cell alterations did modify tumor immunosurveillance, as the adoptive transfer of splenocytes from ApcMin/+ animals into a chemically induced CRC model resulted in the inability to prevent epithelial dysplasia. These results suggest an altered T-cell balance in ApcMin/+ mice may disrupt intestinal homeostasis, consequently limiting intestinal tumor immunosurveillance.


Subject(s)
Colorectal Neoplasms/immunology , Lymph Nodes/pathology , Peyer's Patches/pathology , T-Lymphocytes/immunology , Adenomatous Polyposis Coli Protein/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Polarity , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Granzymes/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Intestinal Mucosa/metabolism , Lymph Nodes/metabolism , Mesentery/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Peyer's Patches/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
2.
Pediatr Diabetes ; 16(6): 407-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25952017

ABSTRACT

A new emphasis has been put on the role of the gastrointestinal (GI) ecosystem in autoimmune diseases; however, there is limited knowledge about its role in type 1 diabetes (T1D). Distinct differences have been observed in intestinal permeability, epithelial barrier function, commensal microbiota, and mucosal innate and adaptive immunity of patients and animals with T1D, when compared with healthy controls. The non-obese diabetic (NOD) mouse and the BioBreeding diabetes prone (BBdp) rat are the most commonly used models to study T1D pathogenesis. With the increasing awareness of the importance of the GI ecosystem in systemic disease, it is critical to understand the basics, as well as the similarities and differences between rat and mouse models and human patients. This review examines the current knowledge of the role of the GI ecosystem in T1D and indicates the extensive opportunities for further investigation that could lead to biomarkers and therapeutic interventions for disease prevention and/or modulation.


Subject(s)
Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Adaptive Immunity , Animals , Autoimmunity , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/physiopathology
3.
Microbiome ; 3: 17, 2015.
Article in English | MEDLINE | ID: mdl-25969735

ABSTRACT

BACKGROUND: Current research has led to the appreciation that there are differences in the commensal microbiota between healthy individuals and individuals that are predisposed to disease. Treatments to reverse disease pathogenesis through the manipulation of the gastrointestinal (GI) microbiota are now being explored. Normalizing microbiota between different strains of mice in the same study is also needed to better understand disease pathogenesis. Current approaches require repeated delivery of bacteria and large numbers of animals and vary in treatment start time. A method is needed that can shift the microbiota of predisposed individuals to a healthy microbiota at an early age and sustain this shift through the lifetime of the individual. RESULTS: We tested cross-fostering of pups within 48 h of birth as a means to permanently shift the microbiota from birth. Taxonomical analysis revealed that the nursing mother was the critical factor in determining bacterial colonization, instead of the birth mother. Data was evaluated using bacterial 16S rDNA sequences from fecal pellets and sequencing was performed on an Illumina Miseq using a 251 bp paired-end library. CONCLUSIONS: The results show that cross-fostering is an effective means to induce an early and maintained shift in the commensal microbiota. This will allow for the evaluation of a prolonged microbial shift and its effects on disease pathogenesis. Cross-fostering will also eliminate variation within control models by normalizing the commensal microbiota between different strains of mice.

4.
J Histochem Cytochem ; 62(4): 237-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24453191

ABSTRACT

Infant formula and breastfeeding are environmental factors that influence the incidence of Type 1 Diabetes (T1D) as well as the acidity of newborn diets. To determine if altering the intestinal microbiome is one mechanism through which an acidic liquid plays a role in T1D, we placed non-obese diabetic (NOD)/ShiLtJt mice on neutral (N) or acidified H2O and monitored the impact on microbial composition and diabetes incidence. NOD-N mice showed an increased development of diabetes, while exhibiting a decrease in Firmicutes and an increase in Bacteroidetes, Actinobacteria, and Proteobacteria from as early as 2 weeks of age. NOD-N mice had a decrease in the levels of Foxp3 expression in CD4(+)Foxp3(+) cells, as well as decreased CD4(+)IL17(+) cells, and a lower ratio of IL17/IFNγ CD4+ T-cells. Our data clearly indicates that a change in the acidity of liquids consumed dramatically alters the intestinal microbiome, the presence of protective Th17 and Treg cells, and the incidence of diabetes. This data suggests that early dietary manipulation of intestinal microbiota may be a novel mechanism to delay T1D onset in genetically pre-disposed individuals.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Fresh Water/chemistry , Gastrointestinal Tract/microbiology , Animals , Bacteroides/isolation & purification , CD4-Positive T-Lymphocytes/metabolism , Clostridium/isolation & purification , DNA, Bacterial/analysis , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Drinking , Feces/microbiology , Female , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/immunology , Hydrogen-Ion Concentration , Interleukin-17/metabolism , Lactobacillus/isolation & purification , Mice , Mice, Inbred NOD
5.
Transpl Immunol ; 28(2-3): 112-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334064

ABSTRACT

Bone marrow reconstitution is utilized as a tool for disease treatment and as a research technique to elucidate the function of bone marrow derived cells. Clinically successful engraftment is indicated by the development of a functioning immune repertoire. In research, reconstitution is considered successful if >85% of splenic leukocytes are of donor origins. Previous work suggests that splenic reconstitution may not be indicative of reconstitution in the mucosa. We sought to evaluate mucosal reconstitution in animals following a standard bone marrow eradication and reconstitution technique. Bone marrow was harvested from adult B6.SJL donor mice (CD45.1) and injected via either the retro-orbital or intraperitoneal route into lethally irradiated B6 (CD45.2) adult or neonatal recipients respectively. The expression of CD45 by flow cytometry was used to calculate reconstitution with respect to immune compartment and cell type. In reconstituted adult animals 93.2±1.5% of splenic leukocytes expressed the donor CD45.1 antigen thus meeting the standard definition of reconstitution, however only 58.6±13.6% of intestinal lamina propria lymphocytes and 52.4±16.0% of intestinal intraepithelial lymphocytes were of donor origin, confirming splenic reconstitution fails to represent peripheral immune reconstitution. T-cells in the gastrointestinal tract are the most poorly reconstituted, while B-cells appear to be almost universally replaced by donor cells. The inadequate mucosal reconstitution was not corrected by evaluating later time points or by performing the bone marrow transfer during the neonatal period. This demonstration that substantial host T-cells remain in the intestinal mucosa after a "successful" bone marrow transplantation should cause a re-evaluation of data from research bone marrow chimera experiments, as well as the mechanisms for complications after clinical bone marrow transplantation.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow/immunology , Cell Lineage , Graft vs Host Disease/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , B-Lymphocytes/cytology , B-Lymphocytes/radiation effects , Bone Marrow/radiation effects , Bone Marrow Transplantation , Mice , T-Lymphocytes/cytology , T-Lymphocytes/radiation effects , Whole-Body Irradiation
6.
Radiat Res ; 178(3): 207-16, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22780103

ABSTRACT

P-glycoprotein (P-gp) has been reported to increase stem cell proliferation and regulate apoptosis. Absence of P-gp results in decreased repair of intestinal epithelial cells after chemical injury. To further explore the mechanisms involved in the effects of P-gp on intestinal injury and repair, we used the well-characterized radiation injury model. In this model, injury repair is mediated by production of prostaglandins (PGE(2)) and lipopolysaccharide (LPS) has been shown to confer radioprotection. B6.mdr1a(-/-) mice and wild-type controls were subjected to 12 Gy total body X-ray irradiation and surviving crypts in the proximal jejunum and distal colon were evaluated 3.5 days after irradiation. B6.mdr1a(-/-) mice exhibited normal baseline stem cell proliferation and COX dependent crypt regeneration after irradiation. However, radiation induced apoptosis was increased and LPS-induced radioprotection was blunted in the C57BL6.mdr1a(-/-) distal colon, compared to B6 wild-type controls. The LPS treatment induced gene expression of the radioprotective cytokine IL-1α, in B6 wild-type controls but not in B6.mdr1a(-/-) animals. Lipopolysaccharid-induced radioprotection was absent in IL-1R1(-/-) animals, indicating a role for IL-1α in radioprotection, and demonstrating that P-gp deficiency interferes with IL-1α gene expression in response to systemic exposure to LPS.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Intestines/injuries , Intestines/radiation effects , Lipopolysaccharides/pharmacology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/radiation effects , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Interleukin-1alpha/metabolism , Intestines/drug effects , Intestines/physiopathology , Mice , Mice, Inbred C57BL , Prostaglandin-Endoperoxide Synthases/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Regeneration/drug effects , Regeneration/radiation effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...