Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Toxicol Pathol ; 47(3): 221-234, 2019 04.
Article in English | MEDLINE | ID: mdl-30844339

ABSTRACT

Pathologic evaluation is crucial to the study of medical devices and integral to the Food and Drug Administration and other regulatory entities' assessment of device safety and efficacy. While pathologic analysis is tailored to the type of device, it generally involves at a minimum gross and microscopic evaluation of the medical device and associated tissues. Due to the complex nature of some implanted devices and specific questions posed by sponsors, pathologic evaluation inherently presents many challenges in accurately assessing medical device safety and efficacy. This laboratory's experience in numerous collaborative projects involving veterinary pathologists, biomedical engineers, physicians, and other scientists has led to a set of interrelated assessments to determine pathologic end points as a means to address these challenges and achieve study outcomes. Thorough device evaluation is often accomplished by utilizing traditional paraffin histology, plastic embedding and microground sections, and advanced imaging modalities. Combining these advanced techniques provides an integrative, comprehensive approach to medical device pathology and enhances medical device safety and efficacy assessment.


Subject(s)
Device Approval/standards , Equipment Safety/standards , Equipment and Supplies/standards , Pathology/methods , Animals , Device Approval/legislation & jurisprudence , Equipment and Supplies/adverse effects , Histological Techniques/methods , Histological Techniques/standards , Humans , Models, Animal , United States , United States Food and Drug Administration
2.
Dig Dis Sci ; 62(8): 1977-1984, 2017 08.
Article in English | MEDLINE | ID: mdl-28573506

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) increases the risk of developing colon cancer. This risk is higher in men compared to women, implicating a role for female hormones in the protection against this disease. Studies from our laboratory demonstrated that estradiol (E2) protects against inflammation-associated colon tumor formation when administered following chemical carcinogen and induction of chronic colitis. AIM: This study seeks to better understand the effect of E2 on acute colitis in the presence and absence of estrogen receptor ß (ERß). METHODS: Inflammation was induced by 2,4,6-trinitrobenzenesulfonic acid in wild-type (WT) and ERß knockout (ERßKO) mice implanted with a control or E2-containing pellet and killed 5 days later. Inflammation and injury were scored by a pathologist. Apoptosis and proliferation were assessed by immunohistochemistry. Cytokines were measured by multiplex analysis. RESULTS: E2 treatment reduced inflammation in the middle colon in WT mice and the distal colon in ERßKO mice compared to control mice. WT mice had reduced IL-6, IL-12, IL-17, GM-CSF, IFN-γ, MCP-1, MIP-1α, and TNF-α, and ERßKO had reduced IL-6 and IFN-γ expression in response to E2. Injury scores were lower in E2-treated ERßKO mice compared to control ERßKO mice. ERßKO mice had increased proliferation in the basal third of crypts in the distal colon and decreased apoptosis in the proximal colon. CONCLUSIONS: These data suggest that E2 has differential protective effects against acute colitis in the presence or absence of ERß and provide insight into how E2 may protect against IBD.


Subject(s)
Colitis/drug therapy , Colitis/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Cytokines/analysis , Cytokines/drug effects , Estrogen Receptor beta/analysis , Estrogen Receptor beta/genetics , Female , Immunohistochemistry , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Trinitrobenzenesulfonic Acid
3.
Carcinogenesis ; 37(2): 206-14, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26717997

ABSTRACT

Perturbations in DNA damage, DNA repair, apoptosis and cell proliferation in the base of the crypt where stem cells reside are associated with colorectal cancer (CRC) initiation and progression. Although the transformation of leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5)(+) cells is an extremely efficient route towards initiating small intestinal adenomas, the role of Lgr5(+) cells in CRC pathogenesis has not been well investigated. Therefore, we further characterized the properties of colonic Lgr5(+) cells compared to differentiated cells in Lgr5-EGFP-IRES-creER(T2) knock-in mice at the initiation stage of carcinogen azoxymethane (AOM)-induced tumorigenesis using a quantitative immunofluorescence microscopy approach. At 12 and 24h post-AOM treatment, colonic Lgr5(+) stem cells (GFP(high)) were preferentially damaged by carcinogen, exhibiting a 4.7-fold induction of apoptosis compared to differentiated (GFP(neg)) cells. Furthermore, with respect to DNA repair, O(6)-methylguanine DNA methyltransferase (MGMT) expression was preferentially induced (by 18.5-fold) in GFP(high) cells at 24h post-AOM treatment compared to GFP(neg) differentiated cells. This corresponded with a 4.3-fold increase in cell proliferation in GFP(high) cells. These data suggest that Lgr5(+) stem cells uniquely respond to alkylation-induced DNA damage by upregulating DNA damage repair, apoptosis and cell proliferation compared to differentiated cells in order to maintain genomic integrity. These findings highlight the mechanisms by which colonic Lgr5(+) stem cells respond to cancer-causing environmental factors.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Homeostasis/drug effects , Intestinal Mucosa/cytology , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Carcinogens/toxicity , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , DNA Damage/drug effects , DNA Damage/physiology , DNA Repair/drug effects , DNA Repair/physiology , Disease Models, Animal , Gene Knock-In Techniques , Homeostasis/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mutagens/toxicity , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Stem Cells/pathology
4.
Vet Dermatol ; 27(4): 311-e77, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27225969

ABSTRACT

BACKGROUND: Endogenous ochronosis is caused by a defect in the enzyme homogentisate 1,2-dioxygenase (HGD), which results in abnormal pigment deposition in the skin and urine abnormalities. Ochronosis previously has not been described histologically or ultrastructurally in a domestic animal species. HYPOTHESIS/OBJECTIVES: To describe the clinical, histopathological and ultrastructural findings in a case of aberrant pigmentation in a cat with features that resemble ochronosis. ANIMAL: A 5-year-old, spayed female Domestic short hair cat presented with multiple black cutaneous plaques on the face and progressive lethargy. The cat's urine turned brown when exposed to air. The familial history of the cat was unknown. METHODS: Clinical examination; histopathology, electron microscopy and mass/energy dispersive X-ray spectroscopy of tissues. RESULTS: Septic peritonitis and additional pigment in the spleen, intestine and lymph node were found at postmortem examination. The pigment was determined to be an organic compound and had a similar histological appearance, staining properties, ultrastructure and composition to ochronotic pigment. No mutations were found in exons 3, 6, 8 and 13 of the HGD gene in the cat. CONCLUSIONS AND CLINICAL IMPORTANCE: To the best of the authors' knowledge, this is the first report of a condition resembling ochronosis in a domestic animal species that has been evaluated with histopathology and advanced imaging techniques. It provides an additional differential in cases of aberrant pigmentation in cats.

5.
Br J Nutr ; 106(4): 519-29, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21401974

ABSTRACT

Both fish oil (FO) and curcumin have potential as anti-tumour and anti-inflammatory agents. To further explore their combined effects on dextran sodium sulphate (DSS)-induced colitis, C57BL/6 mice were randomised to four diets (2 × 2 design) differing in fatty acid content with or without curcumin supplementation (FO, FO+2 % curcumin, maize oil (control, MO) or MO+2 % curcumin). Mice were exposed to one or two cycles of DSS in the drinking-water to induce either acute or chronic intestinal inflammation, respectively. FO-fed mice exposed to the single-cycle DSS treatment exhibited the highest mortality (40 %, seventeen of forty-three) compared with MO with the lowest mortality (3 %, one of twenty-nine) (P = 0·0008). Addition of curcumin to MO increased (P = 0·003) mortality to 37 % compared with the control. Consistent with animal survival data, following the one- or two-cycle DSS treatment, both dietary FO and curcumin promoted mucosal injury/ulceration compared with MO. In contrast, compared with other diets, combined FO and curcumin feeding enhanced the resolution of chronic inflammation and suppressed (P < 0·05) a key inflammatory mediator, NF-κB, in the colon mucosa. Mucosal microarray analysis revealed that dietary FO, curcumin and FO plus curcumin combination differentially modulated the expression of genes induced by DSS treatment. These results suggest that dietary lipids and curcumin interact to regulate mucosal homeostasis and the resolution of chronic inflammation in the colon.


Subject(s)
Colitis/diet therapy , Colon/metabolism , Curcumin/therapeutic use , Cytokines/metabolism , Dietary Supplements , Fish Oils/therapeutic use , Gene Expression Regulation , Acute Disease , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chronic Disease , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Curcumin/adverse effects , Cytokines/genetics , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Fish Oils/adverse effects , Gene Expression Profiling , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Irritants/administration & dosage , Irritants/toxicity , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Random Allocation , Survival Analysis
6.
J Heart Lung Transplant ; 40(1): 56-64, 2021 01.
Article in English | MEDLINE | ID: mdl-33339557

ABSTRACT

BACKGROUND: Histology of thrombosis events in left ventricular assist devices (LVADs) may point to differences between the etiology of either ingested or de novo thrombus formation within LVADs. Materials ingested by the pump would have features suggestive of lifting and folding, whereas thrombi formed de novo would have uniform, parallel layers. This study tested this hypothesis in a cohort of explanted HeartWare Ventricular assist devices (HVADs) (Medtronic, Miami Lakes, Florida). METHODS: Histology of thrombi from 59 explanted HVAD pumps were classified as presumed ingested, presumed de novo, or undeterminable on the basis of pre-defined criteria. The apparent size and location of the thrombotic materials were noted. RESULTS: Histologically, all thrombotic materials were either presumed to be ingested (73%; 95 of 130 total histology cassettes examined) or of undeterminable origin (27%; 35 of 130 histology cassettes). Undetermined origin commonly was due to a lack of sufficient material for analysis. The larger materials (>800 mm3) tended to be in the inflow region. The most common finding was smaller thrombotic materials (<150 mm3) within the pump (64%; 38 of 59 HVADs); when these smaller materials were ingested by the pump, they were most often found within the smaller flow pathways within the pump. CONCLUSIONS: Our study suggests that the thrombi within HVAD pumps are commonly ingested materials rather than de novo thrombus formation within the pump. Further research to understand the source of this ingested material and the consideration to mitigate this complication should be considered.


Subject(s)
Heart Failure/therapy , Heart-Assist Devices/adverse effects , Thrombosis/etiology , Equipment Failure , Humans , Retrospective Studies
7.
Front Vet Sci ; 8: 759585, 2021.
Article in English | MEDLINE | ID: mdl-35252412

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked inherited myopathy that causes progressive skeletal and cardiac muscle disease. Heart lesions were described in the earliest DMD reports, and cardiomyopathy is now the leading cause of death. However, diagnostics and treatment for cardiomyopathy have lagged behind those for appendicular and respiratory skeletal muscle disease. Most animal model studies have been done in the mdx mouse, which has a relatively mild form of cardiomyopathy. Dogs with the genetically homologous condition, Golden Retriever muscular dystrophy (GRMD), develop progressive cardiomyopathy analogous to that seen in DMD. Previous descriptive studies of GRMD cardiomyopathy have mostly been limited to selective sampling of the hearts from young dogs. METHODS AND RESULTS: We systematically assessed cardiac lesions in 31 GRMD and carrier dogs aged 3 to 76 months and a separate cohort of 2-10-year-old normal hounds. Both semi-quantitative lesion scoring and quantitation of the cross-sectional area of fibrosis distinguished dogs with GRMD disease from normal dogs. The carriers generally had intermediate involvement but had even greater fibrosis than GRMD dogs. Fatty infiltration was the most prominent feature in some older GRMD dogs. Vascular hypertrophy was increased in GRMD dogs and correlated positively with lesion severity. Purkinje fiber vacuolation was also increased but did not correlate with lesion severity. Histopathologic changes correlated with late gadolinium enhancement on cardiac MRI. CONCLUSION: These features are generally compatible with those of DMD and further validate GRMD as a useful model to study cardiomyopathy pathogenesis and treatment. Additionally, the nature of some degenerative lesions suggests that functional hypoxia or non-thrombotic ischemia may contribute to disease progression.

8.
MethodsX ; 8: 101297, 2021.
Article in English | MEDLINE | ID: mdl-34434817

ABSTRACT

We sought to develop a reversible staining protocol using micro-computed tomography (micro-CT) paired with a radiopaque contrast agent that allows for three-dimensional in situ visualization and characterization of atherosclerotic plaques. Atherosclerotic porcine coronary arteries were dissected from surrounding myocardium and incubated in iohexol at various concentrations and incubation times and then imaged using direct radiography. Line profiles were generated across the artery x-ray to determine effectiveness of the radiopaque contrast agent to penetrate the tissue. Our studies revealed that, to sufficiently delineate tissue constructs, the minimum effective iohexol concentration and incubation time were 240 mgI/mL for 1 hour. Among all groups, 24 hours of de-staining brought radiopacity back to control levels. After iohexol incubation, micro-CT was performed. Our findings demonstrate that extended staining times and a minimum iohexol concentration of 240 mgI/mL are required for effective tissue perfusion, which eliminates the diffusion distribution profile inherent to the ability of the contrast agent to traverse tissue layers.•Iohexol enhances ex vivo micro-CT imaging of atherosclerotic coronary arteries•Iohexol allows for improved tissue segmentation during micro-CT image analysis•Effectiveness of iohexol penetration of the tissue was dependent on concentration and duration of incubation.

9.
Atherosclerosis ; 313: 43-49, 2020 11.
Article in English | MEDLINE | ID: mdl-33022583

ABSTRACT

BACKGROUND AND AIMS: Visualization of arterial lesions in situ can enhance understanding of atherosclerosis progression and potentially improve experimental therapies. Conventional histology methods for assessing atherosclerotic lesions are robust but are destructive and may prevent further tissue analysis. The objective of the current study was to evaluate a novel, nondestructive method for visualization and characterization of atherosclerotic lesions as an alternative or complementary to routine histology. Thus, we tested the hypothesis that micro-computed tomography (micro-CT) paired with an iodine-based radiopaque stain would effectively characterize atherosclerotic plaques in a manner comparable to routine histology while maintaining sample integrity and providing whole-volume data. METHODS: We examined porcine coronary arteries with varying degrees of atherosclerosis, using micro-CT in the absence and presence of iohexol (240 mgI/ml). Following iohexol washout, routine histological assessment of the samples was performed with hematoxylin and eosin and Masson's trichrome. RESULTS: Iohexol staining generated soft tissue delineation and subsequent atherosclerotic plaque assessment via augmented radiopacity, permitting three-dimensional (3D) reconstruction of these lesions, maintaining in situ architecture. Although plaque distribution and arterial wall tissue layers were discernible, micro-CT was incapable of discriminating cell types comprising the plaque. Calcium phosphate deposition was readily located and visualized in 3D space, independent of iohexol. CONCLUSIONS: The results of this study establish micro-CT, combined with a diffusible radiopaque contrast agent, as a powerful imaging modality for visualizing in situ architecture of atherosclerotic plaques. Our findings demonstrate that micro-CT can be used to identify plaque distribution and calcium deposition complementary to routine histological analysis.


Subject(s)
Iodine , Plaque, Atherosclerotic , Animals , Contrast Media , Coronary Vessels/diagnostic imaging , Plaque, Atherosclerotic/diagnostic imaging , Swine , X-Ray Microtomography
10.
PLoS One ; 15(6): e0235006, 2020.
Article in English | MEDLINE | ID: mdl-32559258

ABSTRACT

Distal limb wounds are common injuries sustained by horses and their healing is fraught with complications due to equine anatomy, prevalence of infection, and challenges associated with wound management. Gallium is a semi-metallic element that has been shown to possess antimicrobial properties and aid in wound healing in various preclinical models. The effects of Gallium have not been studied in equine wound healing. Therefore, the objective of this study was to compare healing rates between gallium-treated and untreated wounds of equine distal limbs and to demonstrate the antimicrobial effects of gallium on wounds inoculated with S. aureus. Using an established model of equine wound healing we demonstrated beneficial effects of 0.5% topical gallium maltolate on equine wound healing. Specifically we documented reduced healing times, reduced bioburden, and reduced formation of exuberant granulation tissue in wounds treated with gallium maltolate as compared with untreated wounds. Gallium appeared to exert its beneficial effects via its well-described antimicrobial actions as well as by altering the expression of specific genes known to be involved in wound healing of horses and other animals. Specifically, gallium maltolate appeared to increase expression of transforming growth factor-ß in both infected and un-infected wounds. Further work is needed to document the effects of gallium on naturally occurring equine wounds and to compare the effects of gallium with other wound treatment options. These data, however, suggest that gallium may be an attractive and novel means of improving equine distal limb wound healing.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Horse Diseases/drug therapy , Leg Injuries/drug therapy , Organometallic Compounds/therapeutic use , Pyrones/therapeutic use , Staphylococcal Infections/drug therapy , Administration, Topical , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Load , Cytokines/genetics , Cytokines/metabolism , Horse Diseases/metabolism , Horses , Leg Injuries/metabolism , Leg Injuries/veterinary , Organometallic Compounds/administration & dosage , Pyrones/administration & dosage , Staphylococcal Infections/metabolism , Staphylococcal Infections/veterinary , Wound Healing
11.
J Nutr ; 139(7): 1328-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19458032

ABSTRACT

We recently demonstrated that (n-3) PUFA trigger the induction of apoptosis in the colon by enhancing phospholipid oxidation and mitochondrial Ca2+ accumulation. To further elucidate the mechanisms regulating oxidative stress-induced apoptosis in vivo, a 2 x 2 experiment was designed using both wild type (control) and manganese-dependent superoxide dismutase (SOD2) heterozygous knockout mice (SOD2(+/-)), which exhibit increased mitochondrial oxidative stress. Mice were fed diets differing only in the type of fat [corn oil or fish oil containing (n-3) PUFA] at 15% by weight for 4 wk. Dietary (n-3) PUFA treatment enhanced (22%) apoptosis in colonic crypts. In addition, SOD2 haploinsufficiency enhanced (20%) apoptosis, which was further increased (36%) by (n-3) PUFA feeding. Dietary lipid source and genotype interactively modulated nitrotyrosine levels (P = 0.027) and inflammation (P = 0.032). These findings demonstrate that the proapoptotic effects of (n-3) PUFA are enhanced in oxidatively stressed SOD2(+/-) mice. Thus, (n-3) PUFA appear to promote an oxidation-reduction imbalance in the intestine, which may directly or indirectly trigger apoptosis and thereby reduce colon cancer risk.


Subject(s)
Apoptosis/drug effects , Colon/enzymology , Fatty Acids, Omega-3/pharmacology , Superoxide Dismutase/deficiency , Animals , Colon/cytology , Colon/drug effects , DNA Primers , Genetic Carrier Screening , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Superoxide Dismutase/genetics
12.
Exp Biol Med (Maywood) ; 243(10): 864-875, 2018 06.
Article in English | MEDLINE | ID: mdl-29874946

ABSTRACT

Inflammatory bowel disease is a complex collection of disorders. Microbial dysbiosis as well as exposure to toxins including xenoestrogens are thought to be risk factors for inflammatory bowel disease development and relapse. Bisphenol-A has been shown to exert estrogenic activity in the colon and alter intestinal function, but the role that xenoestrogens, such as bisphenol-A , play in colonic inflammation has been previously described but with conflicting results. We investigated the ability of bisphenol-A to exacerbate colonic inflammation and alter microbiota metabolites derived from aromatic amino acids in an acute dextran sulfate sodium-induced colitis model. Female C57BL/6 mice were ovariectomized and exposed to bisphenol-A daily for 15 days. Disease activity measures include body weight, fecal consistency, and rectal bleeding. Colons were scored for inflammation, injury, and nodularity. Alterations in the levels of microbiota metabolites derived from aromatic amino acids known to reflect phenotypic changes in the gut microbiome were analyzed. Bisphenol-A exposure increased mortality and worsened disease activity as well as inflammation and nodularity scores in the middle colon region following dextran sulfate sodium exposure. Unique patterns of metabolites were associated with bisphenol-A consumption. Regardless of dextran sulfate sodium treatment, bisphenol-A reduced levels of tryptophan and several metabolites associated with decreased inflammation in the colon. This is the first study to show that bisphenol-A treatment alone can reduce microbiota metabolites derived from aromatic amino acids in the colon which may be associated with increased colonic inflammation and inflammatory bowel disease. Impact statement As rates of inflammatory bowel disease rise, discovery of the mechanisms related to the development of these conditions is important. Environmental exposure is hypothesized to play a role in etiology of the disease, as are alterations in the gut microbiome and the metabolites they produce. This study is the first to show that bisphenol-A alone alters tryptophan and microbiota metabolites derived from aromatic amino acids in a manner consistent with autoimmune diseases, specifically inflammatory bowel diseases, regardless of dextran sulfate sodium treatment. These findings indicate a potential mechanism by which bisphenol-A negatively affects gut physiology to exacerbate inflammation.


Subject(s)
Amino Acids, Aromatic/metabolism , Benzhydryl Compounds/metabolism , Colitis/pathology , Estrogens, Non-Steroidal/metabolism , Gastrointestinal Microbiome/drug effects , Phenols/metabolism , Animals , Benzhydryl Compounds/administration & dosage , Colitis/chemically induced , Colon/pathology , Disease Models, Animal , Estrogens, Non-Steroidal/administration & dosage , Female , Mice, Inbred C57BL , Phenols/administration & dosage , Survival Analysis
13.
Cardiovasc Pathol ; 36: 44-52, 2018.
Article in English | MEDLINE | ID: mdl-30014985

ABSTRACT

Cardiovascular implantable electronic devices (CIEDs) typically incorporate leads that directly contact the endocardium. Post-explant pathology evaluation of formalin-fixed CIED lead implant sites and downstream organs (i.e., lungs) can provide useful safety data to the US Food and Drug Administration; however, current regulatory guidelines do not mandate how the safety data are collected. In this paper, we outline a protocol for preclinical pathology evaluation of leads associated with CIEDs, which includes formalin fixation of the heart and lungs, gross evaluation, and qualitative and quantitative histologic evaluation. We recommend fixation of the whole heart with leads in situ alongside intratracheal formalin infusion; this enables rapid and effective preservation of target tissues and increases histologic quality to allow for accurate qualitative and quantitative pathology evaluation. Overall, we believe that our approach to pathology evaluation of leads may maximize information acquired from preclinical studies, leading to more accurate safety assessments. SUMMARY: This article introduces an established method for pathology evaluation and analysis of cardiac leads recommended for companies and researchers that seek approval from a regulatory body.


Subject(s)
Defibrillators, Implantable/adverse effects , Foreign-Body Reaction/pathology , Lung/pathology , Myocardium/pathology , Pacemaker, Artificial/adverse effects , Tissue Fixation/methods , Animals , Device Removal , Equipment Safety , Fixatives/pharmacology , Foreign-Body Reaction/diagnostic imaging , Formaldehyde/pharmacology , Lung/diagnostic imaging , Microtomy , Models, Animal , Paraffin Embedding , Perfusion , Prosthesis Design , Risk Assessment , X-Ray Microtomography
14.
Cancer Res ; 65(3): 887-97, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705887

ABSTRACT

Low dietary folate intake is associated with an increased risk for colon cancer; however, relevant genetic animal models are lacking. We therefore investigated the effect of targeted ablation of two folate transport genes, folate binding protein 1 (Folbp1) and reduced folate carrier 1 (RFC1), on folate homeostasis to elucidate the molecular mechanisms of folate action on colonocyte cell proliferation, gene expression, and colon carcinogenesis. Targeted deletion of Folbp1 (Folbp1(+/-) and Folbp1(-/-)) significantly reduced (P < 0.05) colonic Folbp1 mRNA, colonic mucosa, and plasma folate concentration. In contrast, subtle changes in folate homeostasis resulted from targeted deletion of RFC1 (RFC1(+/-)). These animals had reduced (P < 0.05) colonic RFC1 mRNA and exhibited a 2-fold reduction in the plasma S-adenosylmethionine/S-adenosylhomocysteine. Folbp1(+/-) and Folbp1(-/-) mice had larger crypts expressed as greater (P < 0.05) numbers of cells per crypt column relative to Folbp1(+/+) mice. Colonic cell proliferation was increased in RFC1(+/-) mice relative to RFC1(+/+) mice. Microarray analysis of colonic mucosa showed distinct changes in gene expression specific to Folbp1 or RFC1 ablation. The effect of folate transporter gene ablation on colon carcinogenesis was evaluated 8 and 38 weeks post-azoxymethane injection in wild-type and heterozygous mice. Relative to RFC1(+/+) mice, RFC1(+/-) mice developed increased (P < 0.05) numbers of aberrant crypt foci at 8 weeks. At 38 weeks, RFC1(+/-) mice developed local inflammatory lesions with or without epithelial dysplasia as well as adenocarcinomas, which were larger relative to RFC1(+/+) mice. In contrast, Folbp1(+/-) mice developed 4-fold (P < 0.05) more lesions relative to Folbp1(+/+) mice. In conclusion, Folbp1 and RFC1 genetically modified mice exhibit distinct changes in colonocyte phenotype and therefore have utility as models to examine the role of folate homeostasis in colon cancer development.


Subject(s)
Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Membrane Transport Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Azoxymethane , Carcinogens , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Cell Cycle/genetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colon/physiology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Folate Receptors, GPI-Anchored , Gene Expression Profiling , Gene Silencing , Genetic Predisposition to Disease , Kidney/metabolism , Kidney/physiology , Male , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Membrane Transport Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylhomocysteine/blood , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/blood , S-Adenosylmethionine/metabolism
15.
Nutrients ; 9(4)2017 Mar 27.
Article in English | MEDLINE | ID: mdl-28346392

ABSTRACT

We have demonstrated that polyphenol-rich sorghum bran diets alter fecal microbiota; however, little is known regarding their effect on colon inflammation. Our aim was to characterize the effect of sorghum bran diets on intestinal homeostasis during dextran sodium sulfate (DSS)-induced colitis. Male Sprague-Dawley rats (N = 20/diet) were provided diets containing 6% fiber from cellulose, or Black (3-deoxyanthocyanins), Sumac (condensed tannins) or Hi Tannin Black (both) sorghum bran. Colitis was induced (N = 10/diet) with three separate 48-h exposures to 3% DSS, and feces were collected. On Day 82, animals were euthanized and the colon resected. Only discrete mucosal lesions, with no diarrhea or bloody stools, were observed in DSS rats. Only bran diets upregulated proliferation and Tff3, Tgfß and short chain fatty acids (SCFA) transporter expression after a DSS challenge. DSS did not significantly affect fecal SCFA concentrations. Bran diets alone upregulated repair mechanisms and SCFA transporter expression, which suggests these polyphenol-rich sorghum brans may suppress some consequences of colitis.


Subject(s)
Colitis/diet therapy , Diet , Dietary Fiber/administration & dosage , Sorghum/chemistry , Animals , Apoptosis , Cell Proliferation , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Edible Grain/chemistry , Epithelial Cells/metabolism , Fatty Acids, Volatile/genetics , Fatty Acids, Volatile/metabolism , Feces/chemistry , Intestinal Mucosa/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Polyphenols/administration & dosage , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism
16.
Sci Rep ; 7(1): 14687, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089621

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most frequently used classes of medications in the world, yet they induce an enteropathy that is associated with high morbidity and mortality. A major limitation to better understanding the pathophysiology and diagnosis of this enteropathy is the difficulty of obtaining information about the primary site of injury, namely the distal small intestine. We investigated the utility of using mRNA from exfoliated cells in stool as a means to surveil the distal small intestine in a murine model of NSAID enteropathy. Specifically, we performed RNA-Seq on exfoliated cells found in feces and compared these data to RNA-Seq from both the small intestinal mucosa and colonic mucosa of healthy control mice or those exhibiting NSAID-induced enteropathy. Global gene expression analysis, data intersection, pathway analysis, and computational approaches including linear discriminant analysis (LDA) and sparse canonical correlation analysis (CCA) were used to assess the inter-relatedness of tissue (invasive) and stool (noninvasive) datasets. These analyses revealed that the exfoliated cell transcriptome closely mirrored the transcriptome of the small intestinal mucosa. Thus, the exfoliome may serve as a non-invasive means of detecting and monitoring NSAID enteropathy (and possibly other gastrointestinal mucosal inflammatory diseases).


Subject(s)
Antirheumatic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions/genetics , Feces/cytology , Intestinal Diseases/genetics , Intestinal Mucosa/physiology , Intestine, Small/physiology , Transcriptome/genetics , Animals , Antirheumatic Agents/therapeutic use , Computational Biology , Disease Models, Animal , Female , Humans , Intestinal Diseases/etiology , Mice , Mice, Inbred C57BL , Organ Specificity
17.
ASAIO J ; 62(3): 252-60, 2016.
Article in English | MEDLINE | ID: mdl-26735557

ABSTRACT

Thromboembolism is a common concern in ventricular assist device (VAD) therapy. Precise VAD response to pass-through thromboembolism needs to be studied in a controlled in vitro setting where specific pump parameters (i.e., power consumption, flow rates, impeller RPM) can be monitored while various types of thrombi are introduced. In this article, we describe a method for creating standardized fibrin thrombi that could be introduced into a mock circulatory loop for testing VAD response to thromboembolism. Donor equine blood collected using a sodium citrate was allowed to clot by adding calcium chloride (CaCl2) while a rotating component applied shear forces to the blood. This rotating force was applied at various speeds and at various distances into the blood. Resulting clots showed similar microscopic features to thrombi taken from explanted clinical VADs. Higher RPM of the rotating component and smaller clearances between the rotating component and the blood created clots that closely resembled ante-explant clots found within VADs in vivo. This method is an effective way to create artificial fibrin clots for use in in vitro experiments to test thromboembolism in VADs.


Subject(s)
Disease Models, Animal , Heart-Assist Devices/adverse effects , Thromboembolism , Animals , Horses , Thromboembolism/etiology
18.
Gut Microbes ; 7(3): 246-61, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27007819

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently used classes of medications in the world. Unfortunately, NSAIDs induce an enteropathy associated with high morbidity and mortality. Although the pathophysiology of this condition involves the interaction of the gut epithelium, microbiota, and NSAIDs, the precise mechanisms by which microbiota influence NSAID enteropathy are unclear. One possible mechanism is that the microbiota may attenuate the severity of disease by specific metabolite-mediated regulation of host inflammation and injury. The microbiota-derived tryptophan-metabolite indole is abundant in the healthy mammalian gut and positively influences intestinal health. We thus examined the effects of indole administration on NSAID enteropathy. Mice (n = 5 per group) were treated once daily for 7 days with an NSAID (indomethacin; 5 mg/kg), indole (20 mg/kg), indomethacin plus indole, or vehicle only (control). Outcomes compared among groups included: microscopic pathology; fecal calprotectin concentration; proportion of neutrophils in the spleen and mesenteric lymph nodes; fecal microbiota composition and diversity; small intestinal mucosal transcriptome; and, fecal tryptophan metabolites. Co-administration of indole with indomethacin: significantly reduced mucosal pathology scores, fecal calprotectin concentrations, and neutrophilic infiltration of the spleen and mesenteric lymph nodes induced by indomethacin; modulated NSAID-induced perturbation of the microbiota, fecal metabolites, and inferred metagenome; and, abrogated a pro-inflammatory gene expression profile in the small intestinal mucosa induced by indomethacin. The microbiota-derived metabolite indole attenuated multiple deleterious effects of NSAID enteropathy, including modulating inflammation mediated by innate immune responses and altering indomethacin-induced shift of the microbiota.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents/metabolism , Enteritis/drug therapy , Gastrointestinal Agents/pharmacology , Indoles/metabolism , Indoles/pharmacology , Inflammation/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Bacteria/classification , Bacteria/isolation & purification , Biota , Disease Models, Animal , Enteritis/chemically induced , Feces/chemistry , Feces/microbiology , Gastrointestinal Agents/administration & dosage , Histocytochemistry , Indoles/administration & dosage , Leukocyte L1 Antigen Complex/analysis , Lymph Nodes/pathology , Mice , Neutrophils/immunology , Spleen/pathology , Treatment Outcome
19.
Cell Death Dis ; 7(11): e2460, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27831561

ABSTRACT

The majority of colon tumors are driven by aberrant Wnt signaling in intestinal stem cells, which mediates an efficient route toward initiating intestinal cancer. Natural lipophilic polyphenols and long-chain polyunsaturated fatty acids (PUFAs) generally suppress Wnt- and NF-κB- (nuclear factor-κ light-chain enhancer of activated B-cell) related pathways. However, the effects of these extrinsic agents on colonic leucine-rich repeat-containing G-protein-coupled receptor 5-positive (Lgr5+) stem cells, the cells of origin of colon cancer, have not been documented to date. Therefore, we examined the effect of n-3 PUFA and polyphenol (curcumin) combination on Lgr5+ stem cells during tumor initiation and progression in the colon compared with an n-6 PUFA-enriched control diet. Lgr5-EGFP-IRES-creERT2 knock-in mice were fed diets containing n-6 PUFA (control), n-3 PUFA, n-6 PUFA+curcumin or n-3 PUFA+curcumin for 3 weeks, followed by 6 azoxymethane (AOM) injections, and terminated 17 weeks after the last injection. To further elucidate the effects of the dietary bioactives at the tumor initiation stage, Lgr5+ stem cells were also assessed at 12 and 24 h post AOM injection. Only n-3 PUFA+curcumin feeding reduced nuclear ß-catenin in aberrant crypt foci (by threefold) compared with control at the progression time point. n-3 PUFA+curcumin synergistically increased targeted apoptosis in DNA-damaged Lgr5+ stem cells by 4.5-fold compared with control at 12 h and maximally reduced damaged Lgr5+ stem cells at 24 h, down to the level observed in saline-treated mice. Finally, RNAseq analysis indicated that p53 signaling in Lgr5+ stem cells from mice exposed to AOM was uniquely upregulated only following n-3 PUFA+curcumin cotreatment. These novel findings demonstrate that Lgr5+ stem cells are uniquely responsive to external dietary cues following the induction of DNA damage, providing a therapeutic strategy for eliminating damaged Lgr5+ stem cells to reduce colon cancer initiation.


Subject(s)
Cell Cycle , Colonic Neoplasms/pathology , Diet , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Aberrant Crypt Foci/metabolism , Animals , Apoptosis/drug effects , Azoxymethane , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chemoprevention , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Curcumin/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Fatty Acids, Omega-3 , Fish Oils/pharmacology , Green Fluorescent Proteins/metabolism , Mice , Regeneration/drug effects , Risk Factors , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
20.
Cancer Res ; 75(24): 5392-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26631266

ABSTRACT

p53 has been shown to mediate cancer stem-like cell function by suppressing pluripotency and cellular dedifferentiation. However, there have been no studies to date that have addressed the specific effects of p53 loss in colonic adult stem cells. In this study, we investigated the consequences of conditionally ablating p53 in the highly relevant Lgr5(+) stem cell population on tumor initiation and progression in the colon. In a mouse model of carcinogen (AOM)-induced colon cancer, tamoxifen-inducible Lgr5-driven deletion of p53 reduced apoptosis and increased proliferation of crypt stem cells, but had no effect on tumor incidence or size. Conversely, in a mouse model of colitis-associated cancer, in which mice are exposed to AOM and the potent inflammation inducer DSS, stem cell-specific p53 deletion greatly enhanced tumor size and incidence in the colon. These novel findings suggest that the loss of p53 function in stem cells enables colonic tumor formation only when combined with DNA damage and chronic inflammation. Furthermore, we propose that stem cell targeting approaches are valuable for interrogating prevention and therapeutic strategies that aim to specifically eradicate genetically compromised stem cells.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Neoplastic Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Animals , Colitis/complications , Colon/pathology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Mutant Strains , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL