Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Mod Pathol ; 35(3): 319-325, 2022 03.
Article in English | MEDLINE | ID: mdl-34628480

ABSTRACT

Optical imaging modalities are emerging as digital microscopy tools for tissue examination. The investigation of these techniques for potential applications in anatomic pathology practice has focused primarily on surgical pathology and has not included cytopathological specimens. We evaluated the feasibility of using digital confocal microscopy (CM) to examine cytopathological specimens. Smears and cell suspensions collected in RPMI solution were prepared from tissue scrapes obtained from surgical resections of breast, lung, liver, and kidney. Air-dried smears and cell pellets obtained from centrifugation of the cell suspensions were stained with 0.6 mM acridine orange and imaged with a CM platform. After completion of imaging, the smears were stained with Diff-Quik (DQ), and cell pellets were routinely processed, embedded in paraffin wax, cut, and stained with hematoxylin and eosin (H&E). We evaluated the mean time to acquire digital CM images; quality of images based on the extent of tissue recognition (0%, grade 0; 1-19%, grade 1; 20-50%, grade 2; >50%, grade 3); comparison of images with DQ- and H&E-stained specimens; and ability to make specific diagnoses. We imaged 91 smears and 52 cell pellets and acquired digital CM images within 2-3 min, with 92% and 88% of images, respectively, from smears and cell pellets showing grade 3 quality. On the basis of CM images, 8 smears (9%) and 7 cell pellets (14%) were categorized as benign, and 83 (91%) and 45 (88%), respectively, as malignant. Specific diagnoses were made by using digital CM images of smears and cell pellets that matched accurately with corresponding DQ- and H&E-stained preparations. The results of our first feasibility study clearly indicated the utility of CM as a next-generation digital microscopy tool for evaluating cytology specimens. Prospective clinical studies are warranted for validating our findings for potential incorporation into cytopathological clinical practice.


Subject(s)
Pathology, Surgical , Feasibility Studies , Hematoxylin , Humans , Microscopy, Confocal/methods , Pathology, Surgical/methods , Prospective Studies
2.
Pharmgenomics Pers Med ; 14: 1565-1574, 2021.
Article in English | MEDLINE | ID: mdl-34898995

ABSTRACT

BACKGROUND: Protein phosphatase 2A (PP2A, a serine/threonine phosphatase) is frequently inactivated in many types of cancer, including primary liver cancer (PLC). Genetic variations in PP2A subunits have been reported to be associated with the risk of many types of cancer but rarely in PLC. This study aims to assess the association between functional polymorphisms of PP2A subunit genes and the risk of PLC in Chinese. METHODS: In a case-control study with a total of 541 PLC patients and 547 controls in Guangxi province of Southern China, we genotyped six putatively functional polymorphisms (rs10421191G>A, rs11453459del>insG, rs1560092T>G, rs7840855C>T, rs1255722G>A and rs10151527A>C) of three PP2A subunit genes (PPP2R1A, PPP2R2A and PPP2R5E) using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry platform. RESULTS: The rs11453459insG variant genotypes (ins/ins+del/ins) of PPP2R1A were found to be significantly associated with an increased risk of PLC compared with the del/del genotype (adjusted OR = 1.290, 95% CI = 1.009-1.650), and the number of insert G allele worked in a dose-dependent manner (P trend= 0.007). The stratified analysis showed that the effects of rs11453459insG variant genotypes were more evident in the subgroup who drink pond-ditch water (adjusted OR = 3.051, 95% CI = 1.264-7.364) than those never drink (P = 0.041). The carriers of rs11453459 del/ins genotype had a significantly lower level of PPP2R1A mRNA expression in liver cancer tissues than those of the del/del genotype (P = 0.021). Furthermore, we used microcystin-LR, a carcinogen presents in the pond-ditch water, to treat human peripheral blood mononuclear cells and found that the cells from carriers of rs11453459insG variant genotypes induced more DNA oxidative damages than those from the del/del genotype carriers (P < 0.001). CONCLUSION: These findings suggest that the PPP2R1A rs11453459del>insG polymorphism is associated with an increased risk of PLC, especially for persons with a history of drinking pond-ditch water. This insertion/deletion polymorphism may be a susceptible biomarker for PLC in Chinese.

3.
PLoS One ; 16(2): e0245334, 2021.
Article in English | MEDLINE | ID: mdl-33571221

ABSTRACT

Providing surgical margin information during breast cancer surgery is crucial for the success of the procedure. The margin is defined as the distance from the tumor to the cut surface of the resection specimen. The consensus among surgeons and radiation oncologists is that there should be no tumor left within 1 to maximum 2 mm from the surface of the surgical specimen. If a positive margin remains, there is substantial risk for tumor recurrence, which may also result in potentially reduced cosmesis and eventual need for mastectomy. In this paper we report a novel multimodal optical imaging instrument based on combined high-resolution confocal microscopy-optical coherence tomography imaging for assessing the presence of potential positive margins on surgical specimens. Since rapid specimen analysis is critical during surgery, this instrument also includes a fluorescence imaging channel to enable rapid identification of the areas of the specimen that have potential positive margins. This is possible by specimen incubation with a cancer specific agent prior to imaging. In this study we used a quenched contrast agent, which is activated by cancer specific enzymes, such as urokinase plasminogen activators (uPA). Using this agent or a similar one, one may limit the use of high-resolution optical imaging to only fluorescence-highlighted areas for visualizing tissue morphology at the sub-cellular scale and confirming or ruling out cancer presence. Preliminary evaluation of this technology was performed on 20 surgical specimens and testing of the optical imaging findings was performed against histopathology. The combination of the three imaging modes allowed for high correlation between optical image analysis and histological ground-truth. The initial results are encouraging, showing instrument capability to assess margins on clinical specimens with a positive predictive value of 1.0 and a negative predictive value of 0.83.


Subject(s)
Breast Neoplasms , Image Processing, Computer-Assisted/methods , Margins of Excision , Microscopy, Confocal , Optical Imaging , Breast/diagnostic imaging , Breast/pathology , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/surgery , Feasibility Studies , Female , Humans , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Optical Imaging/instrumentation , Optical Imaging/methods
4.
JAMA Netw Open ; 3(3): e200476, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32134465

ABSTRACT

Importance: Strategies to procure high-quality core-needle biopsy (CNB) specimens are critical for making basic tissue diagnoses and for ancillary testing. Objectives: To investigate acquisition of fluorescence confocal microscopy (FCM) images of interventional radiology (IR)-guided CNB in real time in the radiology suite and to compare the accuracy of FCM diagnoses with those of hematoxylin-eosin (H&E)-stained CNB sections. Design, Setting, and Participants: In this diagnostic study, FCM imaging of IR-guided CNBs was performed in the radiology suite at a major cancer center for patients with an imaging abnormality from August 1, 2016, to April 30, 2019. The time taken to acquire FCM images and the quality of FCM images based on percentage of interpretable tissue with optimal resolution was recorded. The FCM images were read by 2 pathologists and categorized as nondiagnostic, benign/atypical, or suspicious/malignant; these diagnoses were compared with those made using H&E-stained tissue sections. Cases with discrepant diagnosis were reassessed by the pathologists together for a consensus diagnosis. Data were analyzed from June 3 to July 19, 2019. Interventions: Each IR-guided CNB was stained with 0.6mM acridine orange, subjected to FCM imaging, and then processed to generate H&E-stained sections. Main Outcomes and Measures: Mean time taken for acquisition of FCM images, quality of FCM images based on interpretable percentage of the image, and accuracy of diagnostic categorization based on FCM images compared with H&E-stained sections. Results: A total of 105 patients (57 male [54.3%]; mean [SD] age, 63 [13] years) underwent IR-guided CNBs in a mean (SD) of 7 (2) minutes each. The FCM images showed at least 20% of the tissue with optimal quality in 101 CNB specimens (96.2%). The FCM images were accurately interpreted by the 2 pathologists in 100 of 105 cases (95.2%) (2 false-positive and 3 false-negative) and 90 of 105 cases (85.7%) (6 false-positive and 9 false-negative). A reassessment of 14 discordant diagnoses resulted in consensus diagnoses that were accurate in 101 of 105 cases (96.2%) (1 false-positive and 3 false-negative). Conclusions and Relevance: The ease of acquisition of FCM images of acceptable quality and the high accuracy of the diagnoses suggest that FCM may be useful for rapid evaluation of IR-guided CNBs. This approach warrants further investigation.


Subject(s)
Microscopy, Confocal , Microscopy, Fluorescence , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging , Aged , Biopsy, Large-Core Needle , Female , Fluorescent Dyes , Humans , Image-Guided Biopsy , Male , Middle Aged , Predictive Value of Tests
5.
Arch Pathol Lab Med ; 143(3): 305-313, 2019 03.
Article in English | MEDLINE | ID: mdl-30376375

ABSTRACT

CONTEXT.­: Rapid advances in the fields of biophotonics, computer science, and instrumentation have allowed for high-resolution imaging of biologic tissues. OBJECTIVE.­: To evaluate the quality of images from an optimized confocal fluorescence microscopy (CFM) platform for rapid evaluation of small fragments of tissue, compared with hematoxylin-eosin staining. DESIGN.­: Tissue fragments (up to 1.0 × 0.3 cm) were stained with 0.6 mM acridine orange for 60 seconds and imaged using a CFM platform at 488-nm and 785-nm wavelength. The imaged tissues were then fixed in formalin and processed to generate hematoxylin-eosin-stained tissue sections. The quality of CFM images was scored on a scale of 0 to 3 on the basis of the percentage of the CFM images with recognizable tissue architecture (0, 0%; 1, <20%; 2, 20%-50%; 3, >50%). The diagnoses made using CFM images were compared with those made using histopathologic analysis of the hematoxylin-eosin-stained tissue sections. RESULTS.­: We imaged 118 tissue fragments obtained from 40 breast, 23 lung, 39 kidney, and 16 liver surgical excision specimens. We acquired CFM images in 2 to 3 minutes; 95.8% (113 of 118) of images showed a quality score of 3, and 4.2% (5 of 118) had a score of 2. We achieved a sensitivity of 95.5%, specificity of 97.3%, positive predictive value of 95.5%, and negative predictive value of 97.3%. CONCLUSIONS.­: Our results demonstrate the suitability of the CFM platform for rapid and accurate evaluation of small tissue fragments in surgical pathology practice.


Subject(s)
Image Interpretation, Computer-Assisted/methods , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Pathology, Surgical/methods , Humans , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence/instrumentation , Pathology, Surgical/instrumentation , Sensitivity and Specificity
6.
Clin Cancer Res ; 23(16): 4885-4896, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28490466

ABSTRACT

Purpose: Large diameter perineural prostate cancer is associated with poor outcomes. GDNF, with its coreceptor GFRα1, binds RET and activates downstream pro-oncogenic signaling. Because both GDNF and GFRα1 are secreted by nerves, we examined the role of RET signaling in prostate cancer.Experimental Design: Expression of RET, GDNF, and/or GFRα1 was assessed. The impact of RET signaling on proliferation, invasion and soft agar colony formation, perineural invasion, and growth in vivo was determined. Cellular signaling downstream of RET was examined by Western blotting.Results: RET is expressed in all prostate cancer cell lines. GFRα1 is only expressed in 22Rv1 cells, which is the only line that responds to exogenous GDNF. In contrast, all cell lines respond to GDNF plus GFRα1. Conditioned medium from dorsal root ganglia contains secreted GFRα1 and promotes transformation-related phenotypes, which can be blocked by anti-GFRα1 antibody. Perineural invasion in the dorsal root ganglion assay is inhibited by anti-GFRα antibody and RET knockdown. In vivo, knockdown of RET inhibits tumor growth. RET signaling activates ERK or AKT signaling depending on context, but phosphorylation of p70S6 kinase is markedly increased in all cases. Knockdown of p70S6 kinase markedly decreases RET induced transformed phenotypes. Finally, RET is expressed in 18% of adenocarcinomas and all three small-cell carcinomas examined.Conclusions: RET promotes transformation associated phenotypes, including perineural invasion in prostate cancer via activation of p70S6 kinase. GFRα1, which is secreted by nerves, is a limiting factor for RET signaling, creating a perineural niche where RET signaling can occur. Clin Cancer Res; 23(16); 4885-96. ©2017 AACR.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction/genetics , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins c-ret/metabolism , RNA Interference , RNAi Therapeutics/methods , Tumor Burden/genetics , Xenograft Model Antitumor Assays
7.
J Trauma Acute Care Surg ; 81(2): 358-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27027557

ABSTRACT

BACKGROUND: Intravenous tranexamic acid (TXA) is an effective adjunct after hemorrhagic shock (HS) because of its antifibrinolytic properties. TXA is also a serine protease inhibitor, and recent laboratory data demonstrated that intraluminal TXA into the small bowel inhibited digestive proteases and protected the gut. A Disintegrin And Metalloproteinase 17 (ADAM-17) and tumor necrosis factor α (TNF-α) are effective sheddases of intestinal syndecan-1, which when shed, exposes the underlying intestinal epithelium to digestive proteases and subsequent systemic insult. We therefore hypothesized that intraluminal TXA as a serine protease inhibitor would reduce intestinal sheddases and syndecan-1 shedding, mitigating gut and distant organ (lung) damage. METHODS: Mice underwent 90 minutes of HS to a mean arterial pressure of 35 ± 5 mm Hg followed by the intraluminal administration of TXA or vehicle. After 3 hours, the small intestine, lung, and blood were collected for analysis. RESULTS: Intraluminal TXA significantly reduced gut and lung histopathologic injury and inflammation compared with HS alone. Gut, lung, and systemic ADAM-17 and TNF-α were significantly increased by HS but lessened by TXA. In addition, gut and lung syndecan-1 immunostaining were preserved and systemic shedding lessened after TXA. TXA reduced ADAM-17 and TNF-α, but not syndecan-1, in TXA-sham animals compared with sham vehicles. CONCLUSION: Results of the present study demonstrate a beneficial effect of intraluminal TXA in the gut and lung after experimental HS in part because of the inhibition of the syndecan-1 shedding by ADAM-17 and TNF-α. Further studies are needed to determine if orally administered TXA could provide similar intestinal protection and thus be of potential benefit to patients with survivable hemorrhage at risk for organ injury. This is particularly relevant in patients or soldiers who may not have access to timely medical care.


Subject(s)
Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Lung Injury/prevention & control , Shock, Hemorrhagic/drug therapy , Tranexamic Acid/pharmacology , ADAM17 Protein/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Inflammation/prevention & control , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestine, Small/enzymology , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Syndecan-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Shock ; 44(5): 452-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26263434

ABSTRACT

We have shown in a rodent model of hemorrhagic shock (HS) that fresh frozen plasma (FFP) reduces lung inflammation and injury that are correlated with restitution of syndecan-1. As the gut is believed to contribute to distant organ injury and inflammation after shock, the current study sought to determine if the protective effects of plasma would extend to the gut and to elucidate the contribution of syndecan-1 to this protective effect. We also examined the potential role of TNFα, and a disintegrin and metalloproteinase (ADAM)-17, both intestinal sheddases of syndecan-1. Wild-type (WT) and syndecan-1 (KO) mice were subjected to HS followed by resuscitation with lactated Ringer's (LR) or FFP and compared with shock alone and shams. Small bowel and blood were obtained after 3  h for analysis of mucosal injury and inflammation and TNFα and ADAM-17 protein expression and activity. After HS, gut injury and inflammation were significantly increased compared with shams. Resuscitation with LR decreased both injury and inflammation that were further lessened by FFP. KO mice displayed worsened gut injury and inflammation after HS compared with WT mice, and LR and FFP equivalently inhibited injury and inflammation. Both systemic and intestinal TNFα and ADAM-17 followed similar trends, with increases after HS, reduction by LR, and a further decrease by FFP in WT but not KO mice. In conclusion, FFP decreased gut injury and inflammation after hemorrhagic shock, an effect that was abrogated in syndecan-1 mice. Plasma also decreased TNFα and ADAM-17, representing a potential mechanistic link to its protection via syndecan-1.


Subject(s)
Intestinal Diseases/prevention & control , Plasma , Shock, Hemorrhagic/therapy , Syndecan-1/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Disease Models, Animal , Enteritis/etiology , Enteritis/metabolism , Enteritis/pathology , Enteritis/prevention & control , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Knockout , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/metabolism , Syndecan-1/deficiency , Syndecan-1/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Shock ; 43(4): 327-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25394240

ABSTRACT

We have demonstrated that enteral glutamine provides protection to the postischemic gut, and that peroxisome proliferator-activated receptor-γ (PPARγ) plays a role in this protection. Using Cre/lox technology to generate an intestinal epithelial cell (IEC)-specific PPARγ null mouse model, we now investigated the contribution of IEC PPARγ to glutamine's local and distant organ-protective effects. These mice exhibited absence of expression of PPARγ in the intestine but normal PPARγ expression in other tissues. After 1 h of intestinal ischemia under isoflurane anesthesia, wild-type and null mice received enteral glutamine (60 mM) or vehicle followed by 6 h of reperfusion or 7 days in survival experiments and compared with shams. Small intestine, liver, and lungs were analyzed for injury and inflammatory parameters. Glutamine provided significant protection against gut injury and inflammation, with similar protection in the lung and liver. Changes in systemic tumor necrosis factor-α reflected those seen in the injured organs. Importantly, mice lacking IEC PPARγ had worsened injury and inflammation, and glutamine lost its protective effects in the gut and lung. The survival benefit found in glutamine-treated wild-type mice was not observed in null mice. Using an IEC-targeted loss-of-function approach, these studies provide the first in vivo confirmation in native small intestine and lung that PPARγ is responsible for the protective effects of enteral glutamine in reducing intestinal and lung injury and inflammation and improving survival. These data suggest that early enteral glutamine may be a potential therapeutic modality to reduce shock-induced gut dysfunction and subsequent distant organ injury.


Subject(s)
Epithelial Cells/drug effects , Glutamine/therapeutic use , Intestines/drug effects , PPAR gamma/metabolism , Reperfusion Injury/metabolism , Animals , Glutamine/metabolism , Inflammation , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Ischemia/pathology , Liver/drug effects , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , Shock, Septic/therapy
10.
PLoS One ; 8(9): e76790, 2013.
Article in English | MEDLINE | ID: mdl-24073294

ABSTRACT

BACKGROUND: The role of extracellular signal-regulated protein kinase (ERK) in intestinal ischemia/reperfusion (I/R) injury has not been well investigated. The aim of the current study was to examine the effect of inhibition of the ERK pathway in an in vitro and in vivo model of intestinal I/R injury. METHODS: ERK1/2 activity was inhibited using the specific inhibitor, U0126, in intestinal epithelial cells under hypoxia/reoxygenation conditions and in mice subjected to 1 hour of intestinal ischemia followed by 6 hours reperfusion. In vitro, cell proliferation was assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay, apoptosis by DNA fragmentation, and migration using an in vitro model of intestinal wound healing. Cells were also transfected with a p70S6K plasmid and the effects of overexpression similarly analyzed. In vivo, the effects of U0126 on intestinal cell proliferation and apoptosis, intestinal permeability, lung and intestinal neutrophil infiltration and injury, and plasma cytokine levels were measured. Survival was also assessed after U0126. Activity of p70S6 kinase (p70S6K) was measured by Western blot. RESULTS: In vitro, inhibition of ERK1/2 by U0126 significantly decreased cell proliferation and migration but enhanced cell apoptosis. Overexpression of p70S6K promoted cell proliferation and decreased cell apoptosis. In vivo, U0126 significantly increased cell apoptosis and decreased cell proliferation in the intestine, increased intestinal permeability, intestinal and lung neutrophil infiltration, and injury, as well as systemic pro-inflammatory cytokines, TNF-α, IL-6 and IL-1ß. Mortality was also significantly increased by U0126. Inhibition of ERK1/2 by U0126 also abolished activity of p70S6K both in vitro and in vivo models. CONCLUSION: Pharmacologic inhibition of ERK1/2 by U0126 worsens intestinal IR injury. The detrimental effects are mediated, at least in part, by inhibition of p70S6K, the major effector of mammalian target of rapamycin pathway.


Subject(s)
Butadienes/pharmacology , Intestinal Diseases/pathology , Lung Diseases/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Reperfusion Injury/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Inflammation Mediators/metabolism , Intestinal Diseases/drug therapy , Intestinal Diseases/enzymology , Lung Diseases/drug therapy , Lung Diseases/enzymology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology
11.
PLoS One ; 7(7): e41584, 2012.
Article in English | MEDLINE | ID: mdl-22848534

ABSTRACT

The mTOR signaling pathway plays a crucial role in the regulation of cell growth, proliferation, survival and in directing immune responses. As the intestinal epithelium displays rapid cell growth and differentiation and is an important immune regulatory organ, we hypothesized that mTOR may play an important role in the protection against intestinal ischemia reperfusion (I/R)-induced injury. To better understand the molecular mechanisms by which the mTOR pathway is altered by intestinal I/R, p70S6K, the major effector of the mTOR pathway, was investigated along with the effects of rapamycin, a specific inhibitor of mTOR and an immunosuppressant agent used clinically in transplant patients. In vitro experiments using an intestinal epithelial cell line and hypoxia/reoxygenation demonstrated that overexpression of p70S6K promoted cell growth and migration, and decreased cell apoptosis. Inhibition of p70S6K by rapamycin reversed these protective effects. In a mouse model of gut I/R, an increase of p70S6K activity was found by 5 min and remained elevated after 6 h of reperfusion. Inhibition of p70S6K by rapamycin worsened gut injury, promoted inflammation, and enhanced intestinal permeability. Importantly, rapamycin treated animals had a significantly increased mortality. These novel results demonstrate a key role of p70S6K in protection against I/R injury in the intestine and suggest a potential danger in using mTOR inhibitors in patients at risk for gut hypoperfusion.


Subject(s)
Epithelial Cells/enzymology , Intestinal Diseases/enzymology , Intestinal Mucosa/enzymology , Reperfusion Injury/enzymology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Cell Line , Epithelial Cells/pathology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Intestinal Diseases/pathology , Intestinal Diseases/prevention & control , Intestinal Mucosa/pathology , Male , Mice , Rats , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Sirolimus/adverse effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors
12.
Shock ; 38(1): 57-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22706022

ABSTRACT

Syndecan 1 is the predominant heparan sulfate proteoglycan found on the surface of epithelial cells and, like glutamine, is essential in maintaining the intestinal epithelial barrier. We therefore hypothesized that loss of epithelial syndecan 1 would abrogate the gut-protective effects of enteral glutamine. Both an in vitro and in vivo model of gut ischemia-reperfusion (IR) was utilized. In vitro, intestinal epithelial cells underwent hypoxia-reoxygenation to mimic gut IR with 2 mM (physiologic) or 10 mM glutamine supplementation. Permeability, caspase activity, cell growth, and cell surface and shed syndecan 1 were assessed. In vivo, wild-type and syndecan 1 knockout (KO) mice received ± enteral glutamine followed by gut IR. Intestinal injury was assessed by fluorescent dye clearance and histopathology, permeability as mucosal-to-serosal clearance ex vivo in everted sacs, and inflammation by myeloperoxidase (MPO) activity. In an in vitro model of gut IR, glutamine supplementation reduced epithelial cell permeability and apoptosis and enhanced cell growth. Shed syndecan 1 was reduced by glutamine without an increase in syndecan 1 mRNA. In vivo, intestinal permeability, inflammation, and injury were increased after gut IR in wild-type mice and further increased in syndecan 1 KO mice. Glutamine's attenuation of IR-induced intestinal hyperpermeability, inflammation, and injury was abolished in syndecan 1 KO mice. These results suggest that syndecan 1 plays a novel role in the protective effects of enteral glutamine in the postischemic gut.


Subject(s)
Glutamine/therapeutic use , Intestinal Diseases/prevention & control , Reperfusion Injury/prevention & control , Syndecan-1/physiology , Animals , Caspases/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Hypoxia/physiology , Cells, Cultured , Epithelial Cells/drug effects , Glutamine/pharmacology , Intestinal Diseases/pathology , Intestinal Diseases/physiopathology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestines/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability/drug effects , Rats , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Syndecan-1/deficiency , Syndecan-1/metabolism
13.
Mol Cell Biochem ; 362(1-2): 7-13, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22038625

ABSTRACT

We have previously shown in the post ischemic gut that enteral arginine enhanced injury and inflammation via c-Jun/AP-1 and abrogated peroxisome proliferator-activated receptor (PPAR) γ activity. In the current study, we investigated the mechanism by which arginine inhibited PPARγ in vitro in rat small bowel epithelial IEC-6 cells. Arginine repressed PPARγ transcriptional activity in a time and dose-dependent fashion. Furthermore, downregulation of PPARγ by arginine involved phosphorylation of c-Jun that occurred before to changes in PPARγ transcriptional activity. Silencing of c-Jun increased PPARγ beyond that of nonsilenced cells and was not mitigated by arginine. Using a series of blocking studies, we found no relationship between arginine and the ligand-dependent binding site of PPARγ. In conclusion, arginine decreased PPARγ transcriptional activity in small bowel intestinal epithelial cells. These changes are due, in part, to phosphorylation of c-Jun and may explain the deleterious effects of enteral arginine in the post ischemic gut.


Subject(s)
Arginine/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Transcription, Genetic/drug effects , Animals , Arginine/metabolism , Binding Sites , Cell Line , Epithelial Cells/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering , Rats
14.
Cancer ; 118(13): 3433-45, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22139798

ABSTRACT

BACKGROUND: Patients with chronic myelogenous leukemia (CML) in blast crisis have a poor response to tyrosine kinase inhibitors designed to inhibit the breakpoint cluster region-v-Abelson murine leukemia viral oncogene homolog 1 (BCR-ABL1) oncogene. Recent work has demonstrated that heme oxygenase 1 (HO-1) expression is increased in BCR-ABL1-expressing cells and that the inhibition of HO-1 in CML leads to reduced cellular growth, suggesting that HO-1 may be a plausible target for therapy. The objective of the current study was to clarify the mechanism of HO-1 overexpression and the role of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase as a contributor to this mechanism in CML. METHODS: HO-1 expression was evaluated in bone marrow specimens from patients with CML in various stages of disease, in a transplantation-based model for CML, and in CML cell lines. Chemical and genetic inhibition of the NADPH oxidase was carried out in CML cells. RESULTS: Specimens from patients with CML in blast crisis displayed higher levels of HO-1 staining than specimens from patients with CML in chronic or accelerated phase. HO-1 up-regulation in BCR-ABL1-expressing cells was suppressed by diphenyleneiodonium (DPI), a chemical inhibitor of the NADPH oxidase. Targeting the NADPH oxidase through RNA interference (RNAi) to Ras-related C3 botulinum toxin substrate 1 (Rac1), a dominant-negative Rac1 construct or an inhibitor of Rac1 activity also blunted HO-1 protein expression. Moreover, inhibition of the NADPH oxidase by RNAi directed toward the 47-kd cytosolic subunit of Nox (p47phox) similarly abrogated HO-1 levels. CONCLUSIONS: BCR-ABL1 expression up-regulated HO-1, a survival factor for CML cells. This up-regulation was more pronounced in blast crisis CML relative to early stage disease and was mediated by the NADPH oxidase components Rac1 and p47phox. The expression of p47phox was increased in BCR-ABL1-expressing cells.


Subject(s)
Heme Oxygenase-1/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , NADPH Oxidases/metabolism , Animals , Blast Crisis/genetics , Bone Marrow/metabolism , Cell Line, Tumor , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , NADPH Oxidases/antagonists & inhibitors , Neoplasm Transplantation , Onium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Up-Regulation
15.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G547-54, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21737777

ABSTRACT

Glutamine possesses gut-protective effects both clinically and in the laboratory. We have shown in a rodent model of mesenteric ischemia-reperfusion that enteral glutamine increased peroxisome proliferator-activated receptor-γ (PPAR-γ) and was associated with a reduction in mucosal injury and inflammation. The mechanism by which glutamine activates PPAR-γ is unknown, and we hypothesized that it was via a ligand-dependent mechanism. Intestinal epithelial cells, IEC-6, were co-transfected with PPAR-γ response element-luciferase promoter/reporter construct. Cells were pretreated with increasing concentrations of glutamine ± GW9662 (a specific antagonist of PPAR-γ) and analyzed for PPAR-γ response element luciferase activity as an indicator of PPAR-γ activation. PPAR-γ nuclear activity was assessed by electrophoretic mobility shift assay. Cell lysates were subjected to tandem mass spectroscopy for measurement of prostaglandin and lipoxygenase metabolites. A time- and concentration-dependent increase in PPAR-γ transcriptional activity, but not mRNA or protein, was demonstrated. Activity was abrogated by the PPAR-γ inhibitor, GW9662, and changes in activity correlated with PPAR-γ nuclear binding. Glutamine, via degradation to glutamate, activated the metabolic by-products of the lipoxygenase and linoleic acid pathways, 15-S-hydroxyeicosatetraenoic acid and dehydrogenated 13-hydroxyoctaolecadienoic acid, known endogenous PPAR-γ ligands in the small bowel. This novel mechanism may explain the gut-protective effects of enteral glutamine.


Subject(s)
Glutamine/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Intestinal Mucosa/cytology , Linolenic Acids/metabolism , PPAR gamma/metabolism , Animals , Cells, Cultured , PPAR gamma/agonists , Rats
16.
Hum Pathol ; 42(5): 659-68, 2011 May.
Article in English | MEDLINE | ID: mdl-21239045

ABSTRACT

Although evidence has shown that both the mammalian target of rapamycin and ß-catenin are involved in hepatocellular carcinoma, little is known about their relationship in pathogenesis of hepatocellular carcinoma. To investigate the expression of phosphorylated mammalian target of rapamycin and ß-catenin and their prognostic impacts, as well as their relationship in hepatocellular carcinoma, we analyzed 63 human hepatocellular carcinoma samples by immunohistochemistry. Phosphorylated mammalian target of rapamycin cytoplasmic and ß-catenin cytoplasmic/nuclear-positive immunoreactivities were observed in 63.5% (40/63) and 55.6% (35/63) of the hepatocellular carcinoma specimens, respectively. Significant associations were found between cytoplasmic ß-catenin and phosphorylated mammalian target of rapamycin expression and tumor size (both P < .01) and metastasis (P < .01 and P < .05, respectively). In addition, ß-catenin expression in the cytoplasm was closely associated with the expression of phosphorylated mammalian target of rapamycin. To further explore the relationship between mammalian target of rapamycin and ß-catenin, hepatocellular carcinoma HepG2 and Hep3B cells were treated with ß-catenin siRNA and mammalian target of rapamycin inhibitor, rapamycin; and the expression of phosphorylated mammalian target of rapamycin and ß-catenin in cells was then measured by Western blot. The activity of Wnt/ß-catenin signaling pathway was also assessed by luciferase reporter assay. The cell viability and proliferation were evaluated by thiazolyl blue tetrazolium bromide assay and [(3)H]-thymidine incorporation assay. The results showed that the level of ß-catenin protein expression was markedly decreased by rapamycin in HepG2 and Hep3B cells. The reduction of ß-catenin and mammalian target of rapamycin resulted in inhibition of cell viability proliferation, but the combination of reduction of ß-catenin and mammalian target of rapamycin did not show a synergistic effect on the inhibition of cell viability and proliferation in HepG2 and Hep3B cells. In conclusion, the present study showed that, for the first time, mammalian target of rapamycin regulated the expression level of ß-catenin in hepatocellular carcinoma. Both mammalian target of rapamycin and ß-catenin play important roles in the growth, metastasis, and prognosis of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , beta Catenin/metabolism , Adult , Aged , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Down-Regulation , Drug Synergism , Female , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Male , Middle Aged , Phosphorylation , Signal Transduction , Wnt Proteins/metabolism , Young Adult
17.
Mol Cell Biochem ; 347(1-2): 191-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21046201

ABSTRACT

We previously demonstrated that enteral arginine increased c-Jun/activator protein-1 (AP-1) DNA-binding activity and iNOS expression in a rodent model of mesenteric ischemia/reperfusion (I/R). The objective of this study was to specifically investigate the role of AP-1 in arginine's deleterious effect on the postischemic gut. We hypothesized that AP-1 inhibition would mitigate the effects of arginine. Using a rodent model of mesenteric I/R we demonstrated that gut neutrophil infiltration, activity of c-Jun/AP-1, as well as iNOS expression were increased by I/R and further increased by arginine while lessened by inhibition of c-Jun using the pharmacologic c-Jun N-terminal kinase inhibitor, SP600125. Similar results were demonstrated using a cell culture model of oxidant stress in IEC-6 cells. Importantly, effects of SP600125 were comparable to those of c-Jun silencing. Lastly, the specific iNOS inhibitor, 1400W, had no effect on either AP-1 or c-Jun. In conclusion, SP600125 attenuated the activity of c-Jun/AP-1, iNOS expression, and neutrophil infiltration induced by arginine following mesenteric I/R. Our data suggest that AP-1 inhibition mitigates the injurious inflammatory effects of arginine in the postischemic gut. Further investigation into the pathologic role of enteral arginine in the postischemic gut is warranted.


Subject(s)
Anthracenes/pharmacology , Arginine/administration & dosage , Arginine/pharmacology , Intestine, Small/blood supply , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Reperfusion Injury/pathology , Transcription Factor AP-1/antagonists & inhibitors , Amidines/pharmacology , Animals , Benzylamines/pharmacology , DNA/metabolism , Enteral Nutrition , Gene Silencing/drug effects , Inflammation , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/pathology , Nitric Oxide Synthase Type II/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Transcription Factor AP-1/metabolism
18.
Am J Physiol Gastrointest Liver Physiol ; 299(6): G1344-53, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884886

ABSTRACT

Glutamine plays a key role in intestinal growth and maintenance of gut function, and as we have shown protects the postischemic gut (Kozar RA, Scultz SG, Bick RJ, Poindexter BJ, Desoigne R, Weisbrodt NW, Haber MM, Moore FA. Shock 21: 433-437, 2004). However, the precise mechanisms of the gut protective effects of glutamine have not been well elucidated. In the present study, RNA microarray was performed to obtain differentially expressed genes in intestinal epithelial IEC-6 cells following either 2 mM or 10 mM glutamine. The result demonstrated that specificity protein 3 (Sp3) mRNA expression was downregulated 3.1-fold. PCR and Western blot confirmed that Sp3 expression was decreased by glutamine in a time- and dose-dependent fashion. To investigate the role of Sp3, Sp3 gene siRNA silencing was performed and apoptosis was assessed. Silencing of Sp3 demonstrated a significant increase in Bcl-2 and decrease in Bax protein expression, as well as a decrease in caspase-3, -8, and -9 protein expression and activity. The protein expression of apoptosis-related proteins after hypoxia/reoxygenation was similar to that of normoxia and correlated with a decrease in DNA fragmentation. Importantly, the addition of glutamine to Sp3-silenced cells did not further lessen apoptosis, suggesting that Sp3 plays a major role in the inhibitory effect of glutamine on apoptosis. This novel finding may explain in part the gut-protective effects of glutamine.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Glutamine/pharmacology , Intestinal Mucosa/cytology , Sp3 Transcription Factor/metabolism , Animals , Apoptosis/physiology , Caspases/genetics , Caspases/metabolism , Cell Line , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/physiology , Gene Expression Profiling , Gene Silencing , Genes, bcl-2/physiology , RNA, Small Interfering , Rats , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics
19.
Cancer Lett ; 279(2): 163-70, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19268439

ABSTRACT

Lack of sensitive and specific biomarkers is a major reason for the high rate of hepatocellular carcinoma (HCC) related mortality. The aim of this study was to use surface-enhanced laser desorption/ionization-time-of-flight mass spectroscopy (SELDI-TOF-MS) technology to identify potential protein patterns specific for HCC. Eighty-one patients with hepatitis B-related HCC and 80 healthy controls were randomly divided into a training set (48 HCC, 47 controls) and a testing set (33 HCC, 33 controls). Serum proteomic profiles were measured using SELDI-TOF-MS. A classification tree was established by Biomarker Pattern Software. Candidate biomarkers were separated by HPLC and identified by MALDI-MS/MS and database searching. Forty-eight HCC cases, 54 liver cirrhosis cases and 42 healthy people were clinically validated using candidate biomarkers by SELDI-Immunoassay. Two up-regulated protein peaks were automatically chosen as a classification tree in the training set. These biomarkers were identified as thrombin light chain and growth related oncogene-alpha (GRO-alpha). The sensitivity and specificity of this classification tree were 89.6%. The multivariate model using the two biomarkers and AFP resulted in a sensitivity of 91.7% and specificity of 92.7%, which was significantly better than that of alpha-fetoprotein alone. We conclude that thrombin light chain and GRO-alpha are potential biomarkers of HCC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Liver Neoplasms/blood , Proteome/metabolism , Amino Acid Sequence , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/etiology , Chemokine CXCL1/metabolism , Hepatitis B/blood , Hepatitis B/complications , Humans , Liver Cirrhosis/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Male , Molecular Sequence Data , Multivariate Analysis , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombin/metabolism , alpha-Fetoproteins/metabolism
20.
J Biol Chem ; 284(11): 7114-25, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19131339

ABSTRACT

Signaling initiated by the BCR-ABL1 kinase causes chronic myelogenous leukemia (CML). Recently, we reported that expression of Fyn, a Src kinase, is heightened in CML cells and patient specimens and confers in vitro and in vivo proliferative advantages. Fyn is regulated by redox, and because BCR-ABL1 raises intracellular oxidant levels, which have been implicated in CML progression, we explored the molecular regulation of Fyn. Here we identify the transcription factors that drive redox- and BCR-ABL1-dependent Fyn expression. Promoter deletion analysis in 293T, BaF3, BaF3-p210, and K562 cells identified the region essential for basal transcriptional activity. Mutation of Sp1 and Egr1 binding sites within the essential region diminished Fyn promoter activity and identified Egr1 as conferring redox sensitivity. Gel shift and chromatin immunoprecipitation assays confirmed the binding of Sp1 and Egr1 to the promoter fragments. Importantly, knockdown of Sp1 or Egr1 with small interference RNA or inhibition of Sp1 binding by mithramycin A repressed Fyn protein expression. Our work is the first to define transcription factors that are responsible for endogenous, oxidative stress-dependent and BCR-ABL1-dependent Fyn expression.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-fyn/biosynthesis , Response Elements/physiology , Transcription, Genetic/physiology , Up-Regulation/physiology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Fusion Proteins, bcr-abl/genetics , Humans , K562 Cells , Oxidation-Reduction , Proto-Oncogene Proteins c-fyn/genetics , RNA, Small Interfering/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...