Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Histotechnol ; 45(4): 132-147, 2022 12.
Article in English | MEDLINE | ID: mdl-36317862

ABSTRACT

The central tenet of scientific research is the rigorous application of the scientific method to experimental design, analysis, interpretation, and reporting of results. In order to confer validity to a hypothesis, experimental details must be transparent and results must be reproducible. Failure to achieve this minimum indicates a deficiency in rationale, design, and/or execution, necessitating further experimental refinement or hypothesis reformulation. More importantly, rigorous application of the scientific method advances scientific knowledge by enabling others to identify weaknesses or gaps that can be exploited by new ideas or technology that inevitably extend, improve, or refine a hypothesis. Experimental details, described in manuscript materials and methods, are the principal vehicle used to communicate procedures, techniques, and resources necessary for experimental reproducibility. Recent examination of the biomedical literature has shown that many published articles lack sufficiently detailed methodological information to reproduce experiments. There are few broadly established practice guidelines and quality assurance standards in basic biomedical research. The current paper provides a framework of best practices to address the lack of reporting of detailed materials and methods that is pervasive in histological slide-based assays. Our goal is to establish a structured framework that highlights the key factors necessary for thorough collection of metadata and reporting of slide-based assays.


Subject(s)
Biomedical Research , Metadata , Reproducibility of Results , Research Design , Publications
2.
J Histotechnol ; : 1, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36269236
3.
J Histotechnol ; 45(3): 95, 2022 09.
Article in English | MEDLINE | ID: mdl-35850639
4.
J Histotechnol ; 45(2): 55, 2022 06.
Article in English | MEDLINE | ID: mdl-35607833
10.
J Histotechnol ; 43(4): 161, 2020 12.
Article in English | MEDLINE | ID: mdl-33245264
14.
PLoS One ; 10(1): e0117825, 2015.
Article in English | MEDLINE | ID: mdl-25629976

ABSTRACT

Autoimmune diseases arise from the loss of tolerance to self, and because the etiologies of such diseases are largely unknown, symptomatic treatments rely on anti-inflammatory and analgesic agents. Tolerogenic treatments that can reverse disease are preferred, but again, often thwarted by not knowing the responsible auto-antigens (auto-Ags). Hence, a viable alternative to stimulating regulatory T cells (Tregs) is to induce bystander tolerance. Colonization factor antigen I (CFA/I) has been shown to evoke bystander immunity and to hasten Ag-specific Treg development independent of auto-Ag. To translate in treating human autoimmune diseases, the food-based Lactococcus was engineered to express CFA/I fimbriae, and Lactococcus-CFA/I fermented milk fed to arthritic mice proved highly efficacious. Protection occurred via CD39+ Tregs producing TGF-ß and IL-10 to potently suppress TNF-α production and neutrophil influx into the joints. Thus, these data demonstrate the feasibility of oral nutraceuticals for treating arthritis, and potency of protection against arthritis was improved relative to that obtained with Salmonella-CFA/I.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Arthritis, Experimental/drug therapy , Dietary Supplements , Interleukin-10/metabolism , Milk/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Bystander Effect/immunology , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology
15.
J Immunol ; 192(2): 804-16, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337375

ABSTRACT

A Salmonella therapeutic expressing enterotoxigenic Escherichia coli colonization factor Ag I (CFA/I) fimbriae protects against collagen-induced arthritis (CIA) by eliciting two regulatory T cell (Treg) subsets: TGF-ß-producing Foxp3(-)CD39(+)CD4(+) T cells and IL-10-producing Foxp3(+)CD39(+)CD4(+) T cells. However, it is unclear whether CFA/I fimbriae alone are protective and whether other regulatory cytokines are involved, especially in the context for the EBI3-sharing cytokines, Treg-derived IL-35 and APC-derived IL-27, both capable of suppressing Th17 cells and regulating autoimmune diseases. Subsequent evaluation revealed that a single oral dose of purified, soluble CFA/I fimbriae protected against CIA as effectively as did Salmonella-CFA/I and found that Foxp3(+)CD39(+)CD4(+) T cells were the source of secreted IL-35, whereas IL-27 production by CD11c(+) cells was inhibited. Inquiring into their relevance, CFA/I fimbriae-treated IL-27R-deficient (WSX-1(-/-)) mice were equally protected against CIA as were wild-type mice, suggesting a limited role for IL-27. In contrast, CFA/I fimbriae-mediated protection was abated in EBI3(-/-) mice, accompanied by the loss of TGF-ß- and IL-10-producing Tregs. Adoptive transfer of C57BL/6 CD39(+)CD4(+) T cells to EBI3(-/-) mice with concurrent CFA/I plus IL-35 treatment effectively stimulated Tregs suppressing proinflammatory collagen II-specific Th cells. In contrast, recipients cotransferred with C57BL/6 and EBI3(-/-) CD39(+)CD4(+) T cells and treated with CFA/I plus IL-35 were not protected, implicating the importance of endogenous IL-35 for conferring CFA/I-mediated protection. Thus, CFA/I fimbriae stimulate IL-35 required for the coinduction of TGF-ß and IL-10.


Subject(s)
Arthritis, Experimental/immunology , Escherichia coli/immunology , Fimbriae Proteins/immunology , Fimbriae, Bacterial/immunology , Interleukin-27/immunology , Interleukins/immunology , Animals , Antigens, Bacterial/immunology , Antigens, CD/immunology , Arthritis, Experimental/chemically induced , Forkhead Transcription Factors/immunology , Interleukin-10/immunology , Male , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Receptors, Cytokine/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology
16.
J Leukoc Biol ; 92(2): 375-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22636321

ABSTRACT

Human brucellosis exhibits diverse pathological manifestations that can affect almost any organ. In particular, osteoarticular complications are the most common focal manifestation of brucellosis and occur in 40-80% of patients. In immunocompetent mice, Brucella replication is generally restricted to the spleen, liver, and to a lesser extent, LNs, thereby limiting their use for study of focal inflammation often found in brucellosis. Here, we report that nasal, oral, or peritoneal infection of IFN-γ(-/-) mice with WT Brucella melitensis or Brucella abortus results in joint and periarticular tissue inflammation. Histological analysis of the affected joints revealed inflammatory infiltrates and debris within the joint space colocalizing with Brucella antigen. Osteoarthritis, necrosis, periarticular soft tissue inflammation, and substantial brucellae burdens were observed. Oral rifampicin was effective in clearing infection and halting further progression of focal inflammation from infected IFN-γ(-/-) mice, although some symptoms and swelling remained. Elevated IL-1 ß, but not TNF-α, IL-6, or IL-17, was detected in joint homogenates from infected IFN-γ(-/-) mice. Whereas more susceptible to systemic infection, IL-1R(-/-) mice depleted of IFN-γ were more resistant to focal inflammation than WT mice similarly depleted of IFN-γ. Collectively, these results show IFN-γ(-/-) mice represent a potential model for study of focal inflammation attributed to Brucella infection and will allow evaluation of intervention strategies targeting IL-1, IL-1R, or other inflammatory mediators, with the potential to complement antibiotic-based therapies.


Subject(s)
Brucellosis/immunology , Dermatitis/immunology , Inflammation Mediators/physiology , Interferon-gamma/deficiency , Interleukin-1/physiology , Myositis/immunology , Animals , Brucellosis/genetics , Brucellosis/microbiology , Cells, Cultured , Dermatitis/genetics , Dermatitis/microbiology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interferon-gamma/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myositis/genetics , Myositis/microbiology , Rabbits
17.
PLoS One ; 7(4): e36182, 2012.
Article in English | MEDLINE | ID: mdl-22558374

ABSTRACT

Ovalbumin (OVA) genetically fused to protein sigma 1 (pσ1) results in tolerance to both OVA and pσ1. Pσ1 binds in a multi-step fashion, involving both protein- and carbohydrate-based receptors. To assess the relative pσ1 components responsible for inducing tolerance and the importance of its sialic binding domain (SABD) for immunization, modified OVA-pσ1, termed OVA-pσ1(short), was deleted of its SABD, but with its M cell targeting moiety intact, and was found to be immunostimulatory and enhanced CD4(+) and CD8(+) T cell proliferation. When used to nasally immunize mice given with and without cholera toxin (CT) adjuvant, elevated SIgA and serum IgG responses were induced, and OVA-pσ1(s) was more efficient for immunization than native OVA+CT. The immune antibodies (Abs) were derived from elevated Ab-forming cells in the upper respiratory tissues and submaxillary glands and were supported by mixed Th cell responses. Thus, these studies show that pσ1(s) can be fused to vaccines to effectively elicit improved SIgA responses.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , Ovalbumin/immunology , Sequence Deletion , Vaccination/methods , Adjuvants, Immunologic/metabolism , Animals , Antibody Formation/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/genetics , Cholera Toxin/immunology , Epithelium/immunology , HeLa Cells , Humans , Lymphocytes/cytology , Mice , Peyer's Patches/cytology , Protein Structure, Tertiary , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines/immunology
18.
J Immunol ; 187(9): 4654-66, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21967895

ABSTRACT

Oral immunization with a Salmonella vaccine vector expressing enterotoxigenic Escherichia coli colonization factor Ag I (CFA/I) can protect against collagen-induced arthritis (CIA) by dampening IL-17 and IFN-γ via enhanced IL-4, IL-10, and TGF-ß. To identify the responsible regulatory CD4(+) T cells making the host refractory to CIA, Salmonella-CFA/I induced CD39(+)CD4(+) T cells with enhanced apyrase activity relative to Salmonella vector-immunized mice. Adoptive transfer of vaccine-induced CD39(+)CD4(+) T cells into CIA mice conferred complete protection, whereas CD39(-)CD4(+) T cells did not. Subsequent analysis of vaccinated Foxp3-GFP mice revealed the CD39(+) T cells were composed of Foxp3-GFP(-) and Foxp3-GFP(+) subpopulations. Although each adoptively transferred Salmonella-CFA/I-induced Foxp3(-) and Foxp3(+)CD39(+)CD4(+) T cells could protect against CIA, each subset was not as efficacious as total CD39(+)CD4(+) T cells, suggesting their interdependence for optimal protection. Cytokine analysis revealed Foxp3(-) CD39(+)CD4(+) T cells produced TGF-ß, and Foxp3(+)CD39(+)CD4(+) T cells produced IL-10, showing a segregation of function. Moreover, donor Foxp3-GFP(-) CD4(+) T cells converted to Foxp3-GFP(+) CD39(+)CD4(+) T cells in the recipients, showing plasticity of these regulatory T cells. TGF-ß was found to be essential for protection because in vivo TGF-ß neutralization reversed activation of CREB and reduced the development of CD39(+)CD4(+) T cells. Thus, CD39 apyrase-expressing CD4(+) T cells stimulated by Salmonella-CFA/I are composed of TGF-ß-producing Foxp3(-) CD39(+)CD4(+) T cells and support the stimulation of IL-10-producing Foxp3(+) CD39(+)CD4(+) T cells.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Arthritis, Experimental/immunology , CD4-Positive T-Lymphocytes/immunology , Collagen/toxicity , Forkhead Transcription Factors/physiology , Interleukin-10/physiology , Transforming Growth Factor beta/biosynthesis , Animals , Antigens, CD/biosynthesis , Apyrase/biosynthesis , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , CD4-Positive T-Lymphocytes/metabolism , Collagen/administration & dosage , Fimbriae Proteins , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Interleukin-10/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
19.
J Leukoc Biol ; 90(6): 1043-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21693591

ABSTRACT

Human IBD, including UC and Crohn's disease, is characterized by a chronic, relapsing, and remitting condition that exhibits various features of immunological inflammation and affects at least one/1000 people in Western countries. Polyphenol extracts from a variety of plants have been shown to have immunomodulatory and anti-inflammatory effects. In this study, treatment with APP was investigated to ameliorate chemically induced colitis. Oral but not peritoneal administration of APP during colitis induction significantly protected C57BL/6 mice against disease, as evidenced by the lack of weight loss, colonic inflammation, and shortening of the colon. APP administration dampened the mRNA expression of IL-1ß, TNF-α, IL-6, IL-17, IL-22, CXCL9, CXCL10, CXCL11, and IFN-γ in the colons of mice with colitis. APP-mediated protection requires T cells, as protection was abated in Rag-1(-/-) or TCRα(-/-) mice but not in IL-10(-/-), IRF-1(-/-), µMT, or TCRδ(-/-) mice. Administration of APP during colitis to TCRα(-/-) mice actually enhanced proinflammatory cytokine expression, further demonstrating a requirement for TCRαß cells in APP-mediated protection. APP treatment also inhibited CXCR3 expression by TCRαß cells, but not B or NK cells, in the colons of mice with colitis; however, depletion of CD4(+) or CD8(+) T cells alone did not abolish APP-mediated protection. Collectively, these results show that oral administration of APP protects against experimental colitis and diminishes proinflammatory cytokine expression via T cells.


Subject(s)
Colitis/immunology , Cytokines/antagonists & inhibitors , Dextran Sulfate/toxicity , Inflammation Mediators/physiology , Malus/physiology , Polyphenols/physiology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/pathology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Cytokines/biosynthesis , Cytokines/genetics , Dextran Sulfate/administration & dosage , Female , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/chemistry , Male , Malus/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyphenols/chemistry , T-Lymphocyte Subsets/immunology
20.
Eur J Immunol ; 41(2): 313-23, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268002

ABSTRACT

Stimulation of Ag-specific inducible Treg can enhance resolution of autoimmune disease. Conventional methods to induce Treg often require induction of autoimmune disease or subjection to infection. Reovirus adhesin, protein σ1 (pσ1), can successfully facilitate tolerance when fused to a tolerogen. We tested whether myelin oligodendrocyte glycoprotein (MOG) fused to pσ1 (MOG-pσ1) can stimulate Ag-specific Treg. We show that C57BL/6 mice treated nasally with MOG-pσ1 fail to induce MOG-specific Abs and delayed-type hypersensitivity (DTH) responses and resist EAE. Such resistance was attributed to stimulation of Foxp3(+) Treg, as well as Th2 cells. MOG-pσ1's protective capacity was abrogated in IL-10(-/-) mice, but restored when adoptively transferred with MOG-pσ1-induced Treg. As a therapeutic, MOG-pσ1 diminished EAE within 24 h of nasal application, unlike recombinant MOG (rMOG), pσ1, or pσ1+rMOG, implicating the importance of Ag specificity by pσ1-based therapeutics. MOG-pσ1-treated mice showed elevated IL-4, IL-10, and IL-28 production by CD4(+) T cells, unlike rMOG treated or control mice that produced elevated IFN-γ or IL-17, respectively. These data show the feasibility of using pσ1 as a tolerogen platform for Ag-specific tolerance induction and highlight its potential use as an immunotherapeutic for autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Vaccination/methods , Adoptive Transfer , Animals , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Count , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Female , Glycoproteins/immunology , Hypersensitivity, Delayed/immunology , Immune Tolerance/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spinal Cord/cytology , Spinal Cord/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Th2 Cells/immunology , Th2 Cells/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...