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










Publication year range
1.
J Pharm Sci ; 112(9): 2552-2560, 2023 09.
Article in English | MEDLINE | ID: mdl-37482124

ABSTRACT

Leiomyomas, the most common benign neoplasms of the female reproductive tract, currently have limited medical treatment options. Drugs targeting estrogen/progesterone signaling are used, but side effects and limited efficacy in many cases are major limitation of their clinical use. Previous studies from our laboratory and others demonstrated that 2-methoxyestradiol (2-ME) is promising treatment for uterine fibroids. However, its poor bioavailability and rapid degradation hinder its development for clinical use. The objective of this study is to evaluate the in vivo effect of biodegradable and biocompatible 2-ME-loaded polymeric nanoparticles in a patient-derived leiomyoma xenograft mouse model. PEGylated poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles loaded with 2-ME were prepared by nanoprecipitation. Female 6-week age immunodeficient NOG (NOD/Shi-scid/IL-2Rγnull) mice were used. Estrogen-progesterone pellets were implanted subcutaneously. Five days later, patient-derived human fibroid tumors were xenografted bilaterally subcutaneously. Engrafted mice were treated with 2-ME-loaded or blank (control) PEGylated nanoparticles. Nanoparticles were injected intraperitoneally and after 28 days of treatment, tumor volume was measured by caliper following hair removal, and tumors were removed and weighed. Up to 99.1% encapsulation efficiency was achieved, and the in vitro release profile showed minimal burst release, thus confirming the high encapsulation efficiency. In vivo administration of the 2-ME-loaded nanoparticles led to 51% growth inhibition of xenografted tumors compared to controls (P < 0.01). Thus, 2-ME-loaded nanoparticles may represent a novel approach for the treatment of uterine fibroids.


Subject(s)
Leiomyoma , Nanoparticles , Humans , Mice , Female , Animals , 2-Methoxyestradiol/therapeutic use , Progesterone , Heterografts , Mercaptoethanol/therapeutic use , Mice, Inbred NOD , Leiomyoma/drug therapy , Leiomyoma/pathology , Polymers , Polyethylene Glycols , Estrogens
2.
Br J Cancer ; 128(4): 537-548, 2023 02.
Article in English | MEDLINE | ID: mdl-36482184

ABSTRACT

BACKGROUND: Increases in IL-6 by cancer-associated fibroblasts (CAFs) contribute to colon cancer progression, but the mechanisms involved in the increase of this tumor-promoting cytokine are unknown. The aim of this study was to identify novel targets involved in the dysregulation of IL-6 expression by CAFs in colon cancer. METHODS: Colonic normal (N), hyperplastic, tubular adenoma, adenocarcinoma tissues, and tissue-derived myo-/fibroblasts (MFs) were used in these studies. RESULTS: Transcriptomic analysis demonstrated a striking decrease in alcohol dehydrogenase 1B (ADH1B) expression, a gene potentially involved in IL-6 dysregulation in CAFs. ADH1B expression was downregulated in approximately 50% of studied tubular adenomas and all T1-4 colon tumors, but not in hyperplastic polyps. ADH1B metabolizes alcohols, including retinol (RO), and is involved in the generation of all-trans retinoic acid (atRA). LPS-induced IL-6 production was inhibited by either RO or its byproduct atRA in N-MFs, but only atRA was effective in CAFs. Silencing ADH1B in N-MFs significantly upregulated LPS-induced IL-6 similar to those observed in CAFs and lead to the loss of RO inhibitory effect on inducible IL-6 expression. CONCLUSION: Our data identify ADH1B as a novel potential mesenchymal tumor suppressor, which plays a critical role in ADH1B/retinoid-mediated regulation of tumor-promoting IL-6.


Subject(s)
Cancer-Associated Fibroblasts , Colonic Neoplasms , Interleukin-6 , Humans , Alcohol Dehydrogenase , Cancer-Associated Fibroblasts/metabolism , Colonic Neoplasms/pathology , Fibroblasts/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Tretinoin , Vitamin A/metabolism
3.
J Obstet Gynaecol ; 42(6): 2139-2143, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35166183

ABSTRACT

Uterine leiomyomas are complex tumours with limited medical treatment options. Simvastatin is used to treat hypercholesterolaemia and has shown promising effects as a treatment option for leiomyomas. Previously, our group demonstrated a promising effect of simvastatin treatment in a patient-derived xenograft mouse model. Here, we tested the efficacy of simvastatin liposomal nanoparticles (NPs). After bilateral leiomyoma xenograft implantation, mice (N = 12) were divided into three treatment arms: control, simvastatin and simvastatin-loaded liposome NPs (simvastatin-NPs). Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP; however, the results were not significant. Due to low bioavailability and short half-life of simvastatin, liposomal NPs have the potential to enhance drug delivery, however, in this study NP did not provide improvement over simvastatin, but did demonstrate their potential for the delivery of simvastatin.Impact statementWhat is already known on this subject? Simvastatin treatment in a patient-derived xenograft mouse model reduced tumour growth and decreased proliferation.What do the results of this study add? Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP, however, it did not improve the efficacy of simvastatin at reducing tumour growth and proliferation.What are the implications of these findings for clinical practice and/or further research? More studies are needed to optimise the formulation of NPs to further enhance the sustainable delivery of simvastatin.


Subject(s)
Leiomyoma , Nanoparticles , Animals , Disease Models, Animal , Heterografts , Humans , Ki-67 Antigen , Leiomyoma/drug therapy , Leiomyoma/pathology , Liposomes , Mice , Pilot Projects , Simvastatin/pharmacology
4.
J Biol Chem ; 298(1): 101523, 2022 01.
Article in English | MEDLINE | ID: mdl-34953858

ABSTRACT

Oxidative stress, inflammation, and aberrant activation of microglia in the retina are commonly observed in ocular pathologies. In glaucoma or age-related macular degeneration, the chronic activation of microglia affects retinal ganglion cells and photoreceptors, respectively, contributing to gradual vision loss. However, the molecular mechanisms that cause activation of microglia in the retina are not fully understood. Here we show that exposure of retinal pigment epithelial (RPE) cells to chronic low-level oxidative stress induces mitochondrial DNA (mtDNA)-specific damage, and the subsequent translocation of damaged mtDNA to the cytoplasm results in the binding and activation of intracellular DNA receptor Z-DNA-binding protein 1 (ZBP1). Activation of the mtDNA/ZBP1 pathway triggers the expression of proinflammatory markers in RPE cells. In addition, we show that the enhanced release of extracellular vesicles (EVs) containing fragments of mtDNA derived from the apical site of RPE cells induces a proinflammatory phenotype of microglia via activation of ZBP1 signaling. Collectively, our report establishes oxidatively damaged mtDNA as an important signaling molecule with ZBP1 as its intracellular receptor in the development of an inflammatory response in the retina. We propose that this novel mtDNA-mediated autocrine and paracrine mechanism for triggering and maintaining inflammation in the retina may play an important role in ocular pathologies. Therefore, the molecular mechanisms identified in this report are potentially suitable therapeutic targets to ameliorate development of ocular pathologies.


Subject(s)
DNA, Mitochondrial , Microglia , RNA-Binding Proteins , Retinal Pigment Epithelium , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Humans , Inflammation/metabolism , Microglia/metabolism , Oxidative Stress/genetics , RNA-Binding Proteins/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigments/metabolism
5.
Sci Transl Med ; 13(606)2021 08 11.
Article in English | MEDLINE | ID: mdl-34380769

ABSTRACT

Many women risk unintended pregnancy because of medical contraindications or dissatisfaction with contraceptive methods, including real and perceived side effects associated with the use of exogenous hormones. We pursued direct vaginal delivery of sperm-binding monoclonal antibodies (mAbs) that can limit progressive sperm motility in the female reproductive tract as a strategy for effective nonhormonal contraception. Here, motivated by the greater agglutination potencies of polyvalent immunoglobulins but the bioprocessing ease and stability of immunoglobulin G (IgG), we engineered a panel of sperm-binding IgGs with 6 to 10 antigen-binding fragments (Fabs), isolated from a healthy immune-infertile woman against a unique surface antigen universally present on human sperm. These highly multivalent IgGs (HM-IgGs) were at least 10- to 16-fold more potent and faster at agglutinating sperm than the parent IgG while preserving the crystallizable fragment (Fc) of IgG that mediates trapping of individual spermatozoa in mucus. The increased potencies translated into effective (>99.9%) reduction of progressively motile sperm in the sheep vagina using as little as 33 µg of the 10-Fab HM-IgG. HM-IgGs were produced at comparable yields and had identical thermal stability to the parent IgG, with greater homogeneity. HM-IgGs represent not only promising biologics for nonhormonal contraception but also a promising platform for engineering potent multivalent mAbs for other biomedical applications.


Subject(s)
Immunoglobulin G , Sperm Motility , Animals , Contraception , Female , Humans , Immunoglobulin Fab Fragments , Male , Pregnancy , Sheep , Spermatozoa
6.
Cells ; 10(4)2021 04 06.
Article in English | MEDLINE | ID: mdl-33917426

ABSTRACT

The analysis of circulating cell free DNA (ccf-DNA) is an emerging diagnostic tool for the detection and monitoring of tissue injury, disease progression, and potential treatment effects. Currently, most of ccf-DNA in tissue and liquid biopsies is analysed with real-time quantitative PCR (qPCR) that is primer- and template-specific, labour intensive and cost-inefficient. In this report we directly compare the amounts of ccf-DNA in serum of healthy volunteers, and subjects presenting with various stages of lung adenocarcinoma, and survivors of traumatic brain injury using qPCR and quantitative PicoGreen™ fluorescence assay. A significant increase of ccf-DNA in lung adenocarcinoma and traumatic brain injury patients, in comparison to the group of healthy human subjects, was found using both analytical methods. However, the direct correlation between PicoGreen™ fluorescence and qPCR was found only when mitochondrial DNA (mtDNA)-specific primers were used. Further analysis of the location of ccf-DNA indicated that the majority of DNA is located within lumen of extracellular vesicles (EVs) and is easily detected with mtDNA-specific primers. We have concluded that due to the presence of active DNases in the blood, the analysis of DNA within EVs has the potential of providing rapid diagnostic outcomes. Moreover, we speculate that accurate and rapid quantification of ccf-DNA with PicoGreen™ fluorescent probe used as a point of care approach could facilitate immediate assessment and treatment of critically ill patients.


Subject(s)
Brain Injuries, Traumatic/blood , Cell-Free Nucleic Acids/analysis , DNA, Mitochondrial/analysis , Extracellular Vesicles/genetics , Liquid Biopsy , Severity of Illness Index , Adenocarcinoma of Lung/blood , Adenocarcinoma of Lung/genetics , Adult , Aged , Brain Injuries, Traumatic/genetics , DNA, Mitochondrial/blood , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/genetics , Male , Middle Aged , Organic Chemicals/chemistry
7.
Biol Reprod ; 104(2): 317-324, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33300559

ABSTRACT

High unintended pregnancy rates are partially due to lack of effective nonhormonal contraceptives; development of safe, effective topical vaginal methods will address this need. Preclinical product safety and efficacy assessment requires in vivo testing in appropriate models. The sheep is a good model for the evaluation of vaginally delivered products due to its close similarities to humans. The study objective was to develop an ovine model for efficacy testing of female nonhormonal contraceptives that target human sperm. Fresh human semen was pooled from male volunteers. Nonpregnant female Merino sheep were treated with control or vaginal contraceptive product (IgG antibody with action against sperm or nonoxynol-9 [N9]). Pooled semen was added to the sheep vagina and mixed with product and vaginal secretions. Microscopic assessment of samples was performed immediately and progressive motility (PM) of sperm was compared between treatments. Cytokines CXCL8 and IL1B were assessed in vaginal fluid after instillation of human semen. No adverse reactions or elevations in proinflammatory cytokines occurred in response to human semen. N9 produced signs of acute cellular toxicity while there were no cellular changes after IgG treatment. N9 and IgG had dose-related effects with the highest dose achieving complete sperm immobilization (no sperm with PM). Surrogate post-coital testing of vaginally administered contraceptives that target human semen was developed in an ovine model established for vaginal product preclinical testing. This expanded model can aid the development of much needed nonhormonal topical vaginal contraceptives, providing opportunities for rapid iterative drug development prior to costly, time-intensive human testing.


Subject(s)
Contraceptives, Postcoital/pharmacology , Nonoxynol/pharmacology , Vagina , Animals , Contraceptives, Postcoital/administration & dosage , Female , Humans , Male , Nonoxynol/administration & dosage , Sheep , Sperm Motility/drug effects , Spermatozoa/drug effects
8.
Am J Perinatol ; 36(5): 490-497, 2019 04.
Article in English | MEDLINE | ID: mdl-30193385

ABSTRACT

OBJECTIVE: Epidemiological studies suggest that lactation is associated with long-term maternal health benefits. To avoid confounders in human studies, we used a previously characterized murine model to investigate the long-term effect of lactation on both cardiovascular function and adiposity. STUDY DESIGN: After the delivery of the pups, CD-1 female mice were randomly divided into two groups: lactated and nonlactated (NL). Before pregnancy and at 9 months postdelivery, blood pressure was measured using a tail cuff, visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) were assessed by computed tomography (CT), echocardiography was performed using microultrasound, and cholesterol panels and fasting blood glucose were measured. The data were analyzed using Student's t-test (significance at p < 0.05). RESULTS: There were no differences in baseline parameters between the two groups. At 9 months postdelivery, the NL group weighed significantly more (p = 0.03) and demonstrated a significantly lower cardiac output (p = 0.05) and ejection fraction (p = 0.03). The mice in the NL group also had higher VAT (p < 0.01) and SAT percentiles (p = 0.03). Fasting glucose (p = 0.01) and low-density lipoprotein (p = 0.01) were significantly higher in the NL group at 9 months. CONCLUSION: Our results show the benefit of lactation is not just limited to the immediate postpartum period but it also extends into midlife in a murine model.


Subject(s)
Adiposity/physiology , Blood Pressure/physiology , Cardiac Output/physiology , Lactation/physiology , Animals , Echocardiography , Female , Intra-Abdominal Fat/diagnostic imaging , Mice , Mice, Inbred Strains , Models, Animal , Subcutaneous Fat/diagnostic imaging , Tomography, X-Ray Computed
9.
J Immunol ; 193(5): 2218-29, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070848

ABSTRACT

Signaling via programmed death ligand-1 (PD-L1) and PD-L2 is crucial for maintaining peripheral tolerance. CD90(+) myofibroblasts/fibroblasts (CMFs) are major programmed cell death-1 (PD-1) ligand-expressing cells in normal human colonic mucosa. CMFs suppress activated CD4(+) T cell proliferation via PD-1 ligands. It is not known whether signaling through TLRs contribute to the regulation PD-1 ligands on CMFs upon colonic mucosal tolerance. In this study, we demonstrated that stimulation of TLR4 on human CMFs upregulates PD-L1, but not PD-L2, and reinforces CMF-mediated suppression of CD4(+) T cell proliferation and IFN-γ production. TLR4-mediated upregulation of PD-L1 on CMFs involved NF-κB pathways and was JAK2 and MyD88 dependent. MyD88-dependent stimulation of TLR1/2 and TLR5 also upregulated PD-L1 expression on CMFs in culture. PD-L1 expression was drastically decreased in vivo in the colonic mucosa of mice devoid of MyD88. Induction of MyD88 deficiency in CMFs in fibroblast-specific MyD88 conditional knockout mice resulted in a strong increase in a mucosal IFN-γ expression concomitantly with the abrogation of PD-L1 expression in CMFs under homeostasis and epithelial injury induced by dextran sodium sulfate. Together, these data suggest that MyD88-dependent TLR stimulation of CMFs in the normal colonic mucosa may reinforce these cells' anti-inflammatory capacity and thus contribute to the maintenance of mucosal tolerance.


Subject(s)
B7-H1 Antigen/immunology , Colon/immunology , Immune Tolerance/physiology , Intestinal Mucosa/immunology , Thy-1 Antigens/immunology , Toll-Like Receptor 4/immunology , Animals , B7-H1 Antigen/genetics , Colon/cytology , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Intestinal Mucosa/cytology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myofibroblasts/cytology , Myofibroblasts/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Thy-1 Antigens/genetics , Toll-Like Receptor 4/genetics , Up-Regulation/genetics , Up-Regulation/immunology
10.
Sex Transm Dis ; 40(11): 854-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24113407

ABSTRACT

BACKGROUND: Successful development of topical rectal microbicides requires preclinical evaluation in suitable large animal models. Our previous studies have demonstrated the benefits of high-resolution optical coherence tomography (OCT) to visualize subclinical microbicide toxicity in the sheep vagina. In the current study, we evaluated the potential application of colonoscopy and OCT to visualize and quantify the effects of topical products on sheep colorectal tissue, as assessed by advanced imaging techniques. METHODS: Yearling virginal female sheep were treated rectally with a single 8-mL dose of 0.2% benzalkonium chloride (BZK) solution or phosphate-buffered saline control. Imaging was performed before and 30 minutes after treatment. Colonoscopy findings were evaluated based on mucosal disruption. Optical coherence tomography images were graded based on the integrity of the mucosal layer. Biopsies collected after treatment were evaluated by histology for validation of OCT scoring. RESULTS: Mucosal disruption was observed by colonoscopy in BZK-treated animals, whereas none was present in controls. In contrast to colonoscopy, high-resolution in-depth OCT imaging provided visualization of the morphology of the mucosal layer and underlying muscularis, thus enabling detection of microscopic abnormalities. Noninvasive quantification of drug-induced injury after validation of the scoring system (categories 1, 2, 3) showed increased scores after treatment with BZK (P < 0.001), indicating mucosal injury. CONCLUSIONS: High-resolution OCT can be used as highly sensitive tool to evaluate rectal microbicide effects. Because the sheep rectum has both gross and microscopic similarities to the human, this model is a useful addition to current methods of rectal product toxicity.


Subject(s)
Anal Canal/pathology , Anti-Infective Agents, Local/pharmacology , Benzalkonium Compounds/pharmacology , Intestinal Mucosa/pathology , Tomography, Optical Coherence , Animals , Colonoscopy , Disease Models, Animal , Female , Sheep , Vagina/pathology
12.
Gastroenterology ; 140(7): 2019-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376048

ABSTRACT

BACKGROUND & AIMS: Regulatory T (Treg) cells (CD4+ CD25high FoxP3+) regulate mucosal tolerance; their adoptive transfer prevents or reduces symptoms of colitis in mouse models of inflammatory bowel disease. Colonic CD90+ mesenchymal myofibroblasts and fibroblasts (CMFs) are abundant, nonprofessional antigen-presenting cells in the normal human colonic mucosa that suppress proliferation of activated CD4+ effector T cells. We studied CMF suppressive capacity and evaluated the ability of CMF to induce Treg cells. METHODS: Allogeneic cocultures of CD4+ T cells and CMFs, derived from normal mucosa of patients undergoing colectomy for colon cancer or inflamed colonic tissues from patients with ulcerative colitis or Crohn's disease, were used to assess activation of the Treg cells. RESULTS: Coculture of normal CMF with resting or naïve CD4+ T cells led to development of cells with a Treg phenotype; it also induced proliferation of a CD25+ CD127- FoxP3+ T cells, which expressed CTLA-4, interleukin-10, and transforming growth factor-ß and had suppressive activities. In contrast to dendritic cells, normal CMFs required exogenous interleukin-2 to induce proliferation of naturally occurring Treg cells. Induction of Treg cells by normal CMFs required major histocompatibility complex class II and prostaglandin E2. CMFs from patients with inflammatory bowel diseases had reduced capacity to induce active Treg cells and increased capacity to transiently generate CD4+CD25+/- CD127+ T cells that express low levels of FoxP3. CONCLUSIONS: CMFs suppress the immune response in normal colon tissue and might therefore help maintain colonic mucosal tolerance. Alterations in CMF-mediated induction of Treg cells might promote pathogenesis of inflammatory bowel diseases.


Subject(s)
Cell Proliferation , Colon/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Myofibroblasts/immunology , Paracrine Communication , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Coculture Techniques , Colonic Neoplasms/immunology , Dinoprostone/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immune Tolerance , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Leukocyte Common Antigens/metabolism , Phenotype , Signal Transduction , Time Factors , Transforming Growth Factor beta/metabolism
13.
Gastroenterology ; 135(4): 1228-1237, 1237.e1-2, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18760278

ABSTRACT

BACKGROUND & AIMS: A prominent role for inhibitory molecules PD-L1 and PD-L2 in peripheral tolerance has been proposed. However, the phenotype and function of PD-L-expressing cells in human gut remains unclear. Recent studies suggest that colonic myofibroblasts (CMFs) and fibroblasts are important in the switch from acute inflammation to adaptive immunity. In the normal human colon, CMFs represent a distinct population of major histocompatibility complex class II(+) cells involved in the regulation of mucosal CD4(+) T-cell responses. METHODS: PD-L1 and PD-L2 expression on human CMFs was determined using Western blot, fluorescence-activated cell sorter analysis and confocal microscopy. Lymphoproliferation assays and cytokine enzyme-linked immunosorbent assays were used to evaluate the role of B7 costimulators expressed by CMFs with regard to the regulation of preactivated T-helper cell responses. RESULTS: We demonstrate here the expression of PD-L1/2 molecules by normal human CMF and fibroblasts in situ and in culture. Both molecules support suppressive functions of CMFs in the regulation of activated CD4(+) T-helper cell proliferative responses; blocking this interaction reverses the suppressive effect of CMFs on T-cell proliferation and leads to increased production of the major T-cell growth factor, interleukin (IL)-2. PD-L1/2-mediated CMF suppressive functions are mainly due to the inhibition of IL-2 production, because supplementation of the coculture media with exogenous IL-2 led to partial recovery of activated T-cell proliferation. CONCLUSIONS: Our data suggest that stromal myofibroblasts and fibroblasts may limit T-helper cell proliferative activity in the gut and, thus, might play a prominent role in mucosal intestinal tolerance.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Colon/immunology , Fibroblasts/physiology , Immune Tolerance/physiology , Antigens, CD/genetics , Antigens, CD/metabolism , B7-H1 Antigen , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cell Division/drug effects , Cell Division/immunology , Cells, Cultured , Colon/cytology , Fibroblasts/cytology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-2/pharmacology , Phenotype , Programmed Cell Death 1 Ligand 2 Protein , RNA, Small Interfering , Stromal Cells/cytology , Stromal Cells/physiology , Thy-1 Antigens/metabolism
14.
J Immunol ; 178(12): 8097-106, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548648

ABSTRACT

Food poisoning due to staphylococcal enterotoxins A and B (SEA and SEB) affects hundreds of thousands of people annually. SEA and SEB induce massive intestinal cytokine production, which is believed to be the key factor in staphylococcal enterotoxin enteropathy. MHC class II molecules are the major receptors for staphylococcal enterotoxins. We recently demonstrated that normal human subepithelial intestinal myofibroblasts (IMFs) express MHC class II molecules. We hypothesized that IMFs are among the first cells to respond to staphylococcal enterotoxins and contribute to the cytokine production associated with staphylococcal enterotoxin pathogenesis. We demonstrated here that primary cultured IMFs bind staphylococcal enterotoxins in a MHC class II-dependent fashion in vitro. We also demonstrated that staphylococcal enterotoxins can cross a CaCo-2 epithelial monolayer in coculture with IMFs and bind to the MHC class II on IMFs. IMFs responded to SEA, but not SEB, exposure with 3- to 20-fold increases in the production of proinflammatory chemokines (MCP-1, IL-8), cytokines (IL-6), and growth factors (GM-CSF and G-CSF). The SEA induction of the proinflammatory mediators by IMFs resulted from the efficient cross-linking of MHC class II molecules because cross-linking of class II MHC by biotinylated anti-HLA-DR Abs induced similar cytokine patterns. The studies presented here show that MCP-1 is central to the production of other cytokines elicited by SEA in IMFs because its neutralization with specific Abs prevented the expression of IL-6 and IL-8 by IMFs. Thus, MCP-1 may play a leading role in initiation of inflammatory injury associated with staphylococcal enterotoxigenic disease.


Subject(s)
Chemokine CCL2/metabolism , Enterotoxins/immunology , Intestines/immunology , Staphylococcal Infections/immunology , Antibodies/immunology , Biotinylation , Cells, Cultured , Chemotaxis, Leukocyte , Cytokines/metabolism , Enterotoxins/pharmacology , Fibroblasts/drug effects , Fibroblasts/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation/immunology , Intestines/cytology , Intestines/drug effects , Leukocytes, Mononuclear/immunology , Myoblasts/drug effects , Myoblasts/immunology
15.
J Immunol ; 177(9): 5968-79, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056521

ABSTRACT

The human gastrointestinal mucosa is exposed to a diverse normal microflora and dietary Ags and is a common site of entry for pathogens. The mucosal immune system must respond to these diverse signals with either the initiation of immunity or tolerance. APCs are important accessory cells that modulate T cell responses which initiate and maintain adaptive immunity. The ability of APCs to communicate with CD4+ T cells is largely dependent on the expression of class II MHC molecules by the APCs. Using immunohistochemistry, confocal microscopy, and flow cytometry, we demonstrate that alpha-smooth muscle actin(+), CD90+ subepithelial myofibroblasts (stromal cells) constitutively express class II MHC molecules in normal colonic mucosa and that they are distinct from professional APCs such as macrophages and dendritic cells. Primary isolates of human colonic myofibroblasts (CMFs) cultured in vitro were able to stimulate allogeneic CD4+ T cell proliferation. This process was dependent on class II MHC and CD80/86 costimulatory molecule expression by the myofibroblasts. We also demonstrate that CMFs, engineered to express a specific DR4 allele, can process and present human serum albumin to a human serum albumin-specific and DR4 allele-restricted T cell hybridoma. These studies characterize a novel cell phenotype which, due to its strategic location and class II MHC expression, may be involved in capture of Ags that cross the epithelial barrier and present them to lamina propria CD4+ T cells. Thus, human CMFs may be important in regulating local immunity in the colon.


Subject(s)
Antigen-Presenting Cells/immunology , Histocompatibility Antigens Class II/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Myoblasts, Smooth Muscle/immunology , Actins/analysis , Antigen Presentation , B7-1 Antigen/analysis , B7-2 Antigen/analysis , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Coculture Techniques , Colon/chemistry , Colon/cytology , Colon/immunology , Epithelium/chemistry , Epithelium/immunology , Fibroblasts/chemistry , Fibroblasts/immunology , HLA-DR Antigens/analysis , Histocompatibility Antigens Class II/analysis , Humans , Immunohistochemistry , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/immunology , Microscopy, Confocal , Mucous Membrane/immunology , Myoblasts, Smooth Muscle/chemistry , Myoblasts, Smooth Muscle/drug effects , Stromal Cells/chemistry , Stromal Cells/immunology , Thy-1 Antigens/analysis
16.
Clin Cancer Res ; 10(17): 5870-9, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15355919

ABSTRACT

PURPOSE: Recent data support the hypothesis that the inducible isoform of cyclooxygenase (COX-2) plays a role in the early stages of colonic carcinogenesis and that nonsteroidal anti-inflammatory drugs (NSAIDs) retard the development of colon cancer by modulating COX-2. However, the cell types responsible for producing COX-2 in colorectal adenomas remain a subject of controversy. EXPERIMENTAL DESIGN: COX-2 expression in normal colonic mucosa (n = 50), hyperplastic polyps (n = 43), sporadic adenomas (n = 67), and invasive colonic adenocarcinoma (n = 39) was studied in formalin-fixed and paraffin-embedded tissue sections from endoscopy biopsy and colonic resection specimens. Immunohistochemistry (avidin-biotin complex technique with double immunolabeling) was used to identify the phenotypes of COX-2-producing cells. RESULTS: In colorectal adenomas, increased expression of COX-2 was detected and localized to alpha smooth muscle actin ( proportional, variant SMA)-positive subepithelial stromal cells (myofibroblasts) in the periluminal region of the lamina propria in 63 (94%) of 67 cases. In contrast, in normal colonic mucosa and in hyperplastic polyps with intact epithelium, COX-2 expression was found only in macrophages and endothelial cells. In areas in which the surface epithelium was ulcerated in normal mucosa as well as hyperplastic or neoplastic polyps, COX-2 expression was increased in granulation tissue (and present in macrophages, endothelium, and myofibroblasts). In invasive carcinoma, COX-2 expression in myofibroblasts was limited to the adenomatous portion of the tumor and was detected in 62% of cases (n = 39). In addition, focal expression of COX-2 by malignant epithelial cells was observed in 23% of invasive adenocarcinoma. CONCLUSIONS: These results show that increased COX-2 expression in sporadic adenoma of the colon is common and is localized specifically to subepithelial intestinal myofibroblasts. These findings further support the hypothesis that myofibroblasts are important target cells for NSAID-mediated chemoprevention of colorectal cancer.


Subject(s)
Adenoma/enzymology , Colorectal Neoplasms/enzymology , Epithelium/enzymology , Fibroblasts/enzymology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Actins/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Colon/enzymology , Colorectal Neoplasms/pathology , Cyclooxygenase 2 , Female , Humans , Hyperplasia/enzymology , Immunoenzyme Techniques , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestinal Polyps/enzymology , Intestinal Polyps/pathology , Male , Membrane Proteins , Middle Aged , Muscle, Smooth/enzymology , Neoplasm Invasiveness , Stromal Cells/enzymology
17.
Mol Pharmacol ; 65(2): 470-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742690

ABSTRACT

Acetylsalicylic acid (aspirin) is a cyclooxygenase (COX) inhibitor, yet some of its therapeutic effects are thought to derive from mechanisms unrelated to prostaglandin synthesis inhibition. In human intestinal myofibroblasts, aspirin, at therapeutic doses, had the unexpected effect of inducing prolonged COX-2 expression. This induction was especially pronounced when cells were treated with interleukin-1alpha (IL-1) plus aspirin for 24 h. Sodium salicylate, a poor COX inhibitor, likewise enhanced IL-1-mediated COX-2 gene expression whereas 5-aminosalicylic acid (5-ASA) or indomethacin had no effect. The COX-2 transcriptional rate, measured by nuclear runoff analysis and heterogeneous nuclear RNA reverse transcription-polymerase chain reaction, was only modestly elevated by aspirin treatment. In contrast, aspirin treatment dramatically stabilized the COX-2 message. The COX-2 mRNA half-life in IL-1 treated cells was 1 h and was increased in excess of 5 h in IL-1 + aspirin-treated cells. Phosphorylation of p38 MAPK was enhanced in aspirin-treated cells (but not in cells treated with 5-ASA or indomethacin) for up to 24 h after treatment. Inhibition of p38 activity negated aspirin-mediated COX-2 mRNA stabilization and the resultant increase in COX-2 mRNA and protein levels. The modest transcriptional response seen in aspirin treated cells was also abolished by p38 inhibition. We conclude that aspirin enhances COX-2 expression via sustained activation of p38, which results in prolonged stabilization of the COX-2 message and a slightly elevated transcription rate. Aspirin also enhanced steady-state mRNA levels of other IL-1 modulated genes (IL-1beta, IL-6, groalpha, and TNFalpha) that are likewise regulated at the level of message stability via p38 activation.


Subject(s)
Aspirin/pharmacology , Fibroblasts/enzymology , Intestines/enzymology , Isoenzymes/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Prostaglandin-Endoperoxide Synthases/biosynthesis , RNA Stability/drug effects , Transcription, Genetic/drug effects , Cells, Cultured , Cyclooxygenase 2 , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fibroblasts/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Intestines/cytology , Intestines/drug effects , Isoenzymes/genetics , Membrane Proteins , Mitogen-Activated Protein Kinases/genetics , Prostaglandin-Endoperoxide Synthases/genetics , RNA Stability/genetics , Transcription, Genetic/physiology , p38 Mitogen-Activated Protein Kinases
18.
Gastroenterology ; 124(7): 1855-65, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12806619

ABSTRACT

BACKGROUND & AIMS: Intestinal myofibroblasts (IMFs) express cyclooxygenase 2 (COX-2) early on in polyp progression and respond to pro-inflammatory cytokines. Interleukin (IL)-1alpha induces COX-2 expression in IMF via mitogen-activated protein kinase (MAPK), protein kinase C (PKC), and nuclear factor kappa B (NF-kappaB)-dependent pathways. Because NF-kappaB activity can be mediated by PKC activation and reactive oxygen species (ROS) generation, we examined the relationship of these pathways to IL-1alpha-induced COX-2 expression. METHODS: The effects of specific PKC inhibitors and antioxidants on PKC activation, ROS generation, and COX-2 expression were studied. RESULTS: Immunoprecipitation/kinase (IPK) analysis showed that IL-1alpha increased PKC alpha, delta, and zeta activity 4.5-, 3.1-, and 2.6-fold, respectively, within 5 minutes. Single-cell fluorescence microscopy of 2',7'-dichlorofluorescin diacetate (DCF)-loaded cells showed that IL-1alpha increased ROS levels 2-fold within 15 minutes and this increase was inhibited by 10 micromol/L bisindolylymaleimide I (BIS), a pan-specific PKC inhibitor that also inhibits COX-2 expression. Chelerythrine chloride (CC) (0.5 micromol/L) inhibited classic and novel PKC activity, but not PKCzeta, and enhanced IL-1alpha-mediated ROS generation 4.0-fold and COX-2 expression 1.8-fold. The use of a PKCzeta pseudosubstrate prevented IL-1 from increasing ROS greater than control levels and abolished IL-1alpha-induced COX-2 expression. Small inhibitory RNA (siRNA) for PKCzeta confirmed its role in COX-2 expression. Antioxidants inhibited ROS generation and diminished IL-1alpha-induced COX-2 expression by 80%, without affecting PKC activation. Neither the PKC inhibitors nor the antioxidants prevented NF-kappaB-mediated transcription as determined by reporter gene analysis. CONCLUSIONS: PKCzeta and threshold ROS generation are critical for IL-1alpha-induced COX-2 expression and act concomitantly with NF-kappaB translocation in IMF.


Subject(s)
Colon/enzymology , Interleukin-1/pharmacology , Intestinal Mucosa/enzymology , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Protein Kinase C/physiology , Reactive Oxygen Species , Antioxidants/pharmacology , Cells, Cultured , Cyclooxygenase 2 , Humans , Membrane Proteins , NF-kappa B/physiology , Onium Compounds/pharmacology , Protein Kinase C/antagonists & inhibitors , RNA, Small Interfering/physiology
19.
Arch Pathol Lab Med ; 126(7): 829-36, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12088453

ABSTRACT

CONTEXT: Myofibroblasts are distinct cells with characteristics of both smooth muscle cells and fibroblasts. Through their ability to secrete cytokines, chemokines, prostaglandins, growth factors, and matrix components, they are thought to play critical roles in inflammation, growth, repair, and neoplasia. OBJECTIVE: The goal of this study was to identify the distinct cell populations of the lamina propria of normal colon and colorectal polyps. DESIGN: We studied the expression of alpha-smooth muscle actin (alphaSMA), smooth muscle myosin (SMM), desmin, vimentin, and c-kit by intestinal mesenchymal (stromal) cells in the normal colonic mucosa (n = 5), as well as in hyperplastic polyps (n = 5), sporadic colorectal adenomas (n = 47), and adenomas from patients with familial polyposis (n = 36). RESULTS: In the normal colonic mucosa, the pericryptal stromal cells were alphaSMA+, SMM+, desmin-, and vimentin+, defining them as myofibroblasts. In contrast, cells of the muscularis mucosae were alphaSMA+, SMM+, desmin+, and vimentin-, defining them as smooth muscle cells. alpha-Smooth muscle actin also highlighted direct connections between the muscularis mucosae and the pericryptal myofibroblasts, and vimentin immunostaining showed a network of connections between the alphaSMA+ pericryptal myofibroblasts and the alphaSMA- fibroblasts in the interstitium. In all hyperplastic polyps and adenomatous polyps, the interstitial stromal cells (fibroblasts) now also express alphaSMA and form a syncytium of alphaSMA+ networklike connections throughout the lamina propria. Stromal cells of sporadic adenomas demonstrated the same immunohistochemical staining characteristics displayed by adenomas from patients with familial polyposis and by hyperplastic polyps. Conclusions.-These findings indicate that in normal colon, alphaSMA- fibroblasts are the predominant cell type in the lamina propria. However, the pericryptal (subepithelial) stromal cells are a distinct cell type (alphaSMA+ myofibroblast) that is immunophenotypically different from muscularis mucosae smooth muscle cells and are connected to the interstitial, nonpericryptal fibroblasts with which they exist as a network throughout the lamina propria of the normal colon. Furthermore, in both hyperplastic and neoplastic polyps, there are changes in nonpericryptal fibroblasts from vimentin+, alphaSMA-, and SMM- to vimentin+, alphaSMA+, and SMM+; thus, the interstitial fibroblasts are replaced by myofibroblasts. The factors that cause these changes and the origin of the myofibroblasts need to be determined to clarify the biology of colorectal tumorigenesis.


Subject(s)
Adenoma/metabolism , Colon/metabolism , Colorectal Neoplasms/metabolism , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Intestinal Polyps/metabolism , Adenoma/etiology , Adenoma/pathology , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Biomarkers, Tumor/metabolism , Colon/pathology , Colorectal Neoplasms/pathology , Fibroblasts/pathology , Humans , Hyperplasia/pathology , Immunohistochemistry , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/pathology , Intestinal Polyps/pathology , Neoplasm Proteins/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology
20.
Am J Physiol Cell Physiol ; 282(4): C824-34, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11880271

ABSTRACT

Elevated mucosal interleukin-1 (IL-1) levels are frequently seen during acute and chronic intestinal inflammation, and IL-1 neutralization lessens the severity of inflammation. One major effect of IL-1 is the increased release of eicosanoid mediators via induction of cyclooxygenase-2 (COX-2). One site of COX-2-derived prostaglandin synthesis during acute and chronic intestinal inflammation is the intestinal myofibroblast. COX-2 expression has also been documented in these cells in colonic neoplasms. Thus an understanding of the regulation of COX-2 expression in human intestinal myofibroblasts is important. As an initial step toward this goal we have characterized IL-1alpha signaling pathways that induce COX-2 expression in cultured human intestinal myofibroblasts. IL-1 treatment resulted in a dramatic transcriptional induction of COX-2 gene expression. Activation of nuclear factor-kappaB (NF-kappaB), extracellular signal-regulated protein kinase (ERK), p38, and protein kinase C (PKC) signaling pathways was each necessary for optimal COX-2 induction. In contrast to what occurs in other cell types, including other myofibroblasts such as renal mesangial cells, PKC inhibition did not prevent IL-1-induced NF-kappaB or mitogen activated protein kinase/ stress-activated protein kinase activation, suggesting a novel role for PKC isoforms during this process. The stimulatory effects of PKC, NF-kappaB, ERK-1/2, and presumably c-Jun NH(2)-terminal kinase activation were exerted at the transcriptional level, whereas p38 activation resulted in increased stability of the COX-2 message. We conclude that, in intestinal myofibroblasts, IL-1-mediated induction of COX-2 expression is a complex process that requires input from multiple signaling pathways. Each parallel pathway acts in relative autonomy, the sum of their actions culminating in a dramatic increase in COX-2 transcription and message stability.


Subject(s)
Colon/cytology , Fibroblasts/enzymology , Interleukin-1/pharmacology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Cell Line , Colitis/immunology , Colitis/metabolism , Colon/enzymology , Cyclooxygenase 2 , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Membrane Proteins , Mesoderm/cytology , Mesoderm/enzymology , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Protein Kinase C/metabolism , RNA, Messenger/analysis , Stromal Cells/cytology , Stromal Cells/enzymology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...