Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
ACS Nano ; 16(12): 19626-19650, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36453753

ABSTRACT

We postulate that nanoparticles (NPs) for use in therapeutic applications have largely not realized their clinical potential due to an overall inability to use in vitro results to predict NP performance in vivo. The avian embryo and associated chorioallantoic membrane (CAM) has emerged as an in vivo preclinical model that bridges the gap between in vitro and in vivo, enabling rapid screening of NP behavior under physiologically relevant conditions and providing a rapid, accessible, economical, and more ethical means of qualifying nanoparticles for in vivo use. The CAM is highly vascularized and mimics the diverging/converging vasculature of the liver, spleen, and lungs that serve as nanoparticle traps. Intravital imaging of fluorescently labeled NPs injected into the CAM vasculature enables immediate assessment and quantification of nano-bio interactions at the individual NP scale in any tissue of interest that is perfused with a microvasculature. In this review, we highlight how utilization of the avian embryo and its CAM as a preclinical model can be used to understand NP stability in blood and tissues, extravasation, biocompatibility, and NP distribution over time, thereby serving to identify a subset of NPs with the requisite stability and performance to introduce into rodent models and enabling the development of structure-property relationships and NP optimization without the sacrifice of large populations of mice or other rodents. We then review how the chicken embryo and CAM model systems have been used to accelerate the development of NP delivery and imaging agents by allowing direct visualization of targeted (active) and nontargeted (passive) NP binding, internalization, and cargo delivery to individual cells (of relevance for the treatment of leukemia and metastatic cancer) and cellular ensembles (e.g., cancer xenografts of interest for treatment or imaging of cancer tumors). We conclude by showcasing emerging techniques for the utilization of the CAM in future nano-bio studies.


Subject(s)
Leukemia , Nanoparticles , Neoplasms , Chick Embryo , Humans , Mice , Animals
2.
Prostate ; 79(15): 1767-1776, 2019 11.
Article in English | MEDLINE | ID: mdl-31475741

ABSTRACT

BACKGROUND: The development of phenotypic biomarkers to aid the selection of treatment for patients with castrate-resistant prostate cancer (CRPC) is an important priority. Plasma exosomes have excellent potential as real-time biomarkers to characterize the tumor because they are easily accessible in the blood and contain DNA, RNA, and protein from the parent cell. This study aims to investigate the characteristics of putative prostate-specific plasma extracellular vesicle (EV) markers and their relationship with clinical outcomes. METHODS AND PATIENTS: We investigated plasma EVs in a total of 89 patients with prostate cancer (PCa) at different stages of disease progression. EVs were isolated using both precipitation and ultracentrifugation methods; physical characterization was performed using dynamic light scattering, acetylcholinesterase (AChE) activity, and velocity gradients. An immunocapture method was developed for the evaluation of prostate-specific membrane antigen (PSMA)-positive exosomes. Exosomal messenger RNA (mRNA) was quantified using droplet digital polymerase chain reaction for the expression of KLK3 and androgen receptor splice variant 7 (AR-V7) genes, which code prostate-specific antigen (PSA) and AR-V7, respectively. Serum sex steroids were measured using liquid chromatography-tandem mass spectroscopy. RESULTS: Isolated exosomes from patients with CRPC had a smaller hydrodynamic size than those isolated from localized patients with PCa, while AChE activity showed no difference. Moreover, no differences were observed after initiation of androgen deprivation therapy in serial patient samples. Velocity gradients identified that PSMA-positive exosomes occupied a specific fraction of isolated EVs. A total of 35 patients with CRPC had mRNA analyzed from isolated plasma exosomes. Detectable exosomal KLK3 corresponded with higher concomitant serum PSA measurements, as expected (mean, 112.6 vs 26.61 ng/mL; P = .065). Furthermore, detectable levels of AR-V7 mRNA were associated with a shorter time to progression (median, 16.0 vs 28.0 months; P = .0499). Furthermore, detectable exosomal AR-V7 was significantly associated with testosterone levels below the lower limit of quantification (<0.1 nM). CONCLUSIONS: Our results suggest that exosomal AR-V7 is correlated with lower sex steroid levels in CRPC patients with a poorer prognosis. PSMA immunocapture does not appear sufficient to isolate PCa-specific exosomes.


Subject(s)
Extracellular Vesicles/metabolism , Prostatic Neoplasms, Castration-Resistant/diagnosis , Aged , Aged, 80 and over , Biomarkers, Tumor , Disease Progression , Humans , Kallikreins/metabolism , Male , Middle Aged , Phenotype , Progression-Free Survival , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Survival Rate
3.
Clin Cancer Res ; 24(14): 3299-3308, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29618619

ABSTRACT

Purpose: Homozygous deletions play important roles in carcinogenesis. The genome-wide screening for homozygously deleted genes in many different cancer types with a large number of patient specimens representing the tumor heterogeneity has not been done.Experimental Design: We performed integrative analyses of the copy-number profiles of 10,759 patients across 31 cancer types from The Cancer Genome Atlas project.Results: We found that the type-I interferon, α-, and ß-defensin genes were homozygously deleted in 19 cancer types with high frequencies (7%-31%, median = 12%; interquartile range = 10%-16.5%). Patients with homozygous deletion of interferons exhibited significantly shortened overall or disease-free survival time in a number of cancer types, whereas patients with homozygous deletion of defensins did not significantly associate with worse overall or disease-free survival. Gene expression analyses suggested that homozygous deletion of interferon and defensin genes could activate genes involved in oncogenic and cell-cycle pathways but repress other genes involved in immune response pathways, suggesting their roles in promoting tumorigenesis and helping cancer cells evade immune surveillance. Further analysis of the whole exomes of 109 patients with melanoma demonstrated that the homozygous deletion of interferon (P = 0.0029, OR = 11.8) and defensin (P = 0.06, OR = 2.79) genes are significantly associated with resistance to anti-CTLA4 immunotherapy.Conclusions: Our analysis reveals that the homozygous deletion of interferon and defensin genes is prevalent in human cancers, and importantly this feature can be used as a novel prognostic biomarker for immunotherapy resistance. Clin Cancer Res; 24(14); 3299-308. ©2018 AACR.


Subject(s)
Defensins/genetics , Drug Resistance, Neoplasm/genetics , Gene Frequency , Homozygote , Interferon Type I/genetics , Neoplasms/genetics , Sequence Deletion , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Computational Biology/methods , Databases, Genetic , Genomics/methods , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/mortality , Prognosis , Treatment Outcome
4.
Oncotarget ; 8(42): 71512-71519, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069723

ABSTRACT

Albendazole is an anti-helminthic drug that has been shown to exhibit anti-cancer properties, however its activity in head and neck squamous cell cancer (HNSCC) was unknown. Using a series of in vitro assays, we assessed the ability of albendazole to inhibit proliferation in 20 HNSCC cell lines across a range of albendazole doses (1 nM-10 µM). Cell lines that responded to treatment were further examined for cell death, inhibition of migration and cell cycle arrest. Thirteen of fourteen human papillomavirus-negative HNSCC cell lines responded to albendazole, with an average IC50 of 152 nM. In contrast, only 3 of 6 human papillomavirus-positive HNSCC cell lines responded. Albendazole treatment resulted in apoptosis, inhibition of cell migration, cell cycle arrest in the G2/M phase and altered tubulin distribution. Normal control cells were not measurably affected by any dose tested. This study indicates that albendazole acts to inhibit the proliferation of human papillomavirus-negative HNSCC cell lines and thus warrants further study as a potential chemotherapeutic agent for patients suffering from head and neck cancer.

5.
Int J Mol Sci ; 18(10)2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28937653

ABSTRACT

Previous studies indicate that breast cancer cells with high aldehyde dehydrogenase (ALDH) activity and CD44 expression (ALDHhiCD44⁺) contribute to metastasis and therapy resistance, and that ALDH1 correlates with poor outcome in breast cancer patients. The current study hypothesized that ALDH1 functionally contributes to breast cancer metastatic behavior and therapy resistance. Expression of ALDH1A1 or ALDH1A3 was knocked down in MDA-MB-468 and SUM159 human breast cancer cells using siRNA. Resulting impacts on ALDH activity (Aldefluor® assay); metastatic behavior and therapy response in vitro (proliferation/adhesion/migration/colony formation/chemotherapy and radiation) and extravasation/metastasis in vivo (chick choroiallantoic membrane assay) was assessed. Knockdown of ALDH1A3 but not ALDH1A1 in breast cancer cells decreased ALDH activity, and knockdown of ALDH1A1 reduced breast cancer cell metastatic behavior and therapy resistance relative to control (p < 0.05). In contrast, knockdown of ALDH1A3 did not alter proliferation, extravasation, or therapy resistance, but increased adhesion/migration and decreased colony formation/metastasis relative to control (p < 0.05). This is the first study to systematically examine the function of ALDH1 isozymes in individual breast cancer cell behaviors that contribute to metastasis. Our novel results indicate that ALDH1 mediates breast cancer metastatic behavior and therapy resistance, and that different enzyme isoforms within the ALDH1 family differentially impact these cell behaviors.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Aldehyde Oxidoreductases/metabolism , Breast Neoplasms/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehyde Oxidoreductases/genetics , Animals , Breast Neoplasms/genetics , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Chick Embryo , Chickens , Female , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism
6.
Mol Cell Biol ; 36(13): 1836-55, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27141054

ABSTRACT

Antiangiogenic therapies, such as sunitinib, have revolutionized renal cell carcinoma (RCC) treatment. However, a precarious understanding of how resistance emerges and a lack of tractable experimental systems hinder progress. We evaluated the potential of primary RCC cultures (derived from tumors and tumor grafts) to signal to endothelial cells (EC) and fibroblasts in vitro and to stimulate angiogenesis ex vivo in chorioallantoic membrane (CAM) assays. From 65 patients, 27 primary cultures, including several from patients with sunitinib-resistant RCC, were established. RCC cells supported EC survival in coculture assays and induced angiogenesis in CAM assays. RCC-induced EC survival was sensitive to sunitinib in half of the tumors and was refractory in tumors from resistant patients. Sunitinib sensitivity correlated with vascular endothelial growth factor (VEGF) production. RCC induced paracrine extracellular signal-regulated kinase (ERK) activation in EC which was inhibited by sunitinib in sensitive but not in resistant tumors. As determined by fibroblast growth factor receptor substrate 2 (FRS2) phosphorylation in fibroblasts, RCC broadly induced low-level fibroblast growth factor receptor (FGFR) signaling. Whereas ERK activation in EC was uniformly inhibited by combined VEGF/platelet-derived growth factor (PDGF)/FGF receptor inhibitors, paracrine ERK activation in fibroblasts was blocked in only a fraction of tumors. Our data show that RCC activates EC through VEGF-dependent and -independent pathways, that sunitinib sensitivity correlates with VEGF-mediated ERK activation, and that combined inhibition of VEGF/PDGF/FGF receptors is sufficient to inhibit mitogenic signaling in EC but not in fibroblasts.


Subject(s)
Carcinoma, Renal Cell/metabolism , Drug Resistance, Neoplasm , Kidney Neoplasms/metabolism , Paracrine Communication , Receptors, Fibroblast Growth Factor/metabolism , Animals , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Paracrine Communication/drug effects , Pyrroles/pharmacology , Sunitinib , Tumor Cells, Cultured
7.
Cell Mol Life Sci ; 71(24): 4881-94, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24879294

ABSTRACT

Our previous work showed that Cyclin A2 deficiency promotes cell invasion in fibroblasts. Given that the majority of cancers emerge from epithelia, we explored novel functions for Cyclin A2 by depleting it in normal mammary epithelial cells. This caused an epithelial to mesenchymal transition (EMT) associated with loss of cell-to-cell contacts, decreased E-Cadherin expression and increased invasive properties characterized by a reciprocal regulation of RhoA and RhoC activities, where RhoA-decreased activity drove cell invasiveness and E-Cadherin delocalization, and RhoC-increased activity only supported cell motility. Phenotypes induced by Cyclin A2 deficiency were exacerbated upon oncogenic activated-Ras expression, which led to an increased expression of EMT-related transcriptional factors. Moreover, Cyclin A2-depleted cells exhibited stem cell-like properties and increased invasion in an in vivo avian embryo model. Our work supports a model where Cyclin A2 downregulation facilitates cancer cell EMT and metastatic dissemination.


Subject(s)
Cell Movement/genetics , Cyclin A2/genetics , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Communication/genetics , Cell Cycle/genetics , Cell Line , Cell Survival/genetics , Cyclin A2/metabolism , Fibroblasts/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoblotting , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
8.
Methods Mol Biol ; 872: 1-14, 2012.
Article in English | MEDLINE | ID: mdl-22700400

ABSTRACT

Cell migration and metastasis are key features of aggressive tumors. These processes can be difficult to study, as they often occur deep within the body of a cancer patient or an experimental animal. In vitro assays are able to model some aspects of these processes, and a number of assays have been developed to assess cancer cell motility, migration, and invasion. However, in vitro assays have inherent limitations that may miss important aspects of these processes as they occur in vivo. The chick embryo provides a powerful model for studying these processes in vivo, facilitated by the external and accessible nature of the chorioallantoic membrane (CAM), a well-vascularized tissue that surrounds the embryo. When coupled with multiple fluorescent approaches to labeling both cancer cells and the embryonic vasculature, along with image analysis tools, the chick CAM model offers cost-effective, rapid assays for studying cancer cell migration and metastasis in a physiologically-relevant, in vivo setting. Here, we present recent developments of detailed procedures for using shell-less chick embryos, coupled with fluorescent labeling of cancer cells and/or chick vasculature, to study cancer cell migration and metastasis in vivo.


Subject(s)
Cell Movement/physiology , Fluorescence , Animals , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/metabolism
9.
J Vis Exp ; (52)2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21730939

ABSTRACT

Current technologies for tumor imaging, such as ultrasound, MRI, PET and CT, are unable to yield high-resolution images for the assessment of nanoparticle uptake in tumors at the microscopic level(1,2,3,) highlighting the utility of a suitable xenograft model in which to perform detailed uptake analyses. Here, we use high-resolution intravital imaging to evaluate nanoparticle uptake in human tumor xenografts in a modified, shell-less chicken embryo model. The chicken embryo model is particularly well-suited for these in vivo analyses because it supports the growth of human tumors, is relatively inexpensive and does not require anesthetization or surgery 4,5. Tumor cells form fully vascularized xenografts within 7 days when implanted into the chorioallantoic membrane (CAM)( 6). The resulting tumors are visualized by non-invasive real-time, high-resolution imaging that can be maintained for up to 72 hours with little impact on either the host or tumor systems. Nanoparticles with a wide range of sizes and formulations administered distal to the tumor can be visualized and quantified as they flow through the bloodstream, extravasate from leaky tumor vasculature, and accumulate at the tumor site. We describe here the analysis of nanoparticles derived from Cowpea mosaic virus (CPMV) decorated with near-infrared fluorescent dyes and/or polyethylene glycol polymers (PEG) (7, 8, 9,10,11). Upon intravenous administration, these viral nanoparticles are rapidly internalized by endothelial cells, resulting in global labeling of the vasculature both outside and within the tumor(7,12). PEGylation of the viral nanoparticles increases their plasma half-life, extends their time in the circulation, and ultimately enhances their accumulation in tumors via the enhanced permeability and retention (EPR) effect (7, 10,11). The rate and extent of accumulation of nanoparticles in a tumor is measured over time using image analysis software. This technique provides a method to both visualize and quantify nanoparticle dynamics in human tumors.


Subject(s)
Colonic Neoplasms/metabolism , Nanoparticles/administration & dosage , Animals , Chick Embryo , Chorioallantoic Membrane/blood supply , Colonic Neoplasms/blood supply , Comovirus/chemistry , Fluorescent Dyes/chemistry , HT29 Cells , Humans , Image Processing, Computer-Assisted/methods , Microinjections/instrumentation , Microinjections/methods , Microscopy/methods , Nanoparticles/chemistry , Polyethylene Glycols/chemistry
10.
Nat Protoc ; 5(8): 1406-17, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20671724

ABSTRACT

Viral nanoparticles are a novel class of biomolecular agents that take advantage of the natural circulatory and targeting properties of viruses to allow the development of therapeutics, vaccines and imaging tools. We have developed a multivalent nanoparticle platform based on the cowpea mosaic virus (CPMV) that facilitates particle labeling at high density with fluorescent dyes and other functional groups. Compared with other technologies, CPMV-based viral nanoparticles are particularly suited for long-term intravital vascular imaging because of their biocompatibility and retention in the endothelium with minimal side effects. The stable, long-term labeling of the endothelium allows the identification of vasculature undergoing active remodeling in real time. In this study, we describe the synthesis, purification and fluorescent labeling of CPMV nanoparticles, along with their use for imaging of vascular structure and for intravital vascular mapping in developmental and tumor angiogenesis models. Dye-labeled viral nanoparticles can be synthesized and purified in a single day, and imaging studies can be conducted over hours, days or weeks, depending on the application.


Subject(s)
Comovirus/isolation & purification , Embryo, Mammalian/blood supply , Embryo, Nonmammalian/blood supply , Endothelium, Vascular/anatomy & histology , Nanoparticles , Nanotechnology/methods , Neoplasms/blood supply , Animals , Chickens , Embryo Culture Techniques , Fabaceae/virology , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Mice , Microinjections/methods , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods
11.
Transplantation ; 90(1): 4-13, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20463646

ABSTRACT

BACKGROUND: There is increasing evidence for a role for autoimmunity in transplant rejection. It has previously been shown that autoantibodies to vimentin (Vim) accelerate acute rejection of murine cardiac allografts. We have investigated whether autoimmunity to Vim contributes to development of cardiac allograft vasculopathy (CAV). METHODS: Two well-established minor mismatch murine models of CAV were used, transplantation of 129/sv hearts into T-cell-depleted C57Bl/6 (B6) recipients and transplantation of FVB hearts into nonimmunosuppressed DBA/1 recipients. Recipients were immunized with recombinant mouse Vim in complete Freunds adjuvant, and controls received hen egg lysozyme 2 weeks before transplantation. T cell and antibody responses to Vim were assessed by ELISPOT and ELISA, respectively. CAV within transplanted hearts was assessed by quantitative morphometry of occluded vessels, presence of smooth muscle cells, deposition of C3d, and confocal microscopy. RESULTS: Allografts were harvested from B6 recipients at days 30 and 45 and from DBA/1 recipients at days 18 and 35. At all days, there was significantly more intimal occlusion of arteries of Vim -immunized mice than controls. There was significantly more smooth muscle cell alpha actin in vessels from Vim-immunized mice, and more C3d deposited in hearts from Vim-immunized mice. Confocal microscopy demonstrated colocalization of Vim with C3d on endothelial cells, leukocytes, and platelets in allogeneic but not syngeneic hearts. Serum from Vim-immunized mice, but not controls, caused platelet/leukocyte conjugation when added to mouse leukocytes. CONCLUSION: The autoimmune response to Vim accelerates CAV progression in these minor-mismatched models.


Subject(s)
Heart Transplantation/immunology , Transplantation, Homologous/immunology , Transplantation, Isogeneic/immunology , Vimentin/immunology , Animals , Antibodies, Monoclonal/pharmacology , Autoimmunity/immunology , Enzyme-Linked Immunosorbent Assay , Graft Rejection/pathology , Heart Transplantation/pathology , Histocompatibility Testing , Leukocytes/drug effects , Leukocytes/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Confocal , Minor Histocompatibility Antigens/immunology , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Transplantation, Heterotopic/immunology
12.
Am J Pathol ; 170(4): 1415-27, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392180

ABSTRACT

Autoimmune responses to vimentin occur after solid organ transplantation, but their pathogenic effects are unclear. The aim of these studies was to investigate the effects of vimentin preimmunization on allogeneic and isografted hearts in a murine transplant model. Immunization of C57BL/6 mice with murine vimentin in complete Freund's adjuvant resulted in anti-vimentin antibodies and vimentin-reactive Th-1 cells. Transplantation of 129/sv hearts into vimentin-immunized C57BL/6 recipients resulted in accelerated rejection (8.4 +/- 1.5 days; n = 18), compared with hen egg lysozyme-immunized C57BL/6 (13.3 +/- 2.2 days; n = 10; P < 0.0001, log-rank test). In contrast, isografts continued to beat beyond 90 days. Immunohistochemical analysis of allografts from vimentin/complete Freund's adjuvant mice demonstrated increased numbers of T cells and enhanced microvascular deposition of C3d, CD41, and P-selectin compared with controls. Antibodies were necessary for accelerated rejection, shown by the fact that vimentin-immunized B-cell-deficient IgH6 mice did not show accelerated rejection of 129/sv allografts, but rejection was restored by adoptive transfer of serum containing anti-vimentin antibodies. Eluates from donor hearts placed in vimentin/complete Freund's adjuvant recipients contained anti-vimentin antibodies, shown by Western blotting. Confocal imaging of rejected hearts de-monstrated presence of vimentin and C3d on apoptosed leukocytes, endothelial cells, and platelet/leukocyte conjugates. These results demonstrate that autoantibodies to vimentin, in conjunction with the alloimmune response, have a pathogenic role in allograft rejection.


Subject(s)
Autoantibodies/immunology , Graft Rejection/immunology , Heart Transplantation , Vimentin/immunology , Animals , Blotting, Western , CD3 Complex/immunology , CD4 Antigens/immunology , Graft Survival/immunology , Immune Tolerance , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Isoantibodies/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Protein Binding/immunology , T-Lymphocytes/immunology , Transplantation, Homologous , Transplantation, Isogeneic , Vaccination/methods
13.
Cardiovasc Pathol ; 15(2): 63-74, 2006.
Article in English | MEDLINE | ID: mdl-16533694

ABSTRACT

BACKGROUND: Coxsackievirus B3 (CVB3) is the major causative agent of myocarditis in humans. In the mouse model, the inflammatory phase of myocarditis results in extensive damage to the heart and triggers profound extracellular matrix (ECM) remodeling, which may ultimately lead to dilated cardiomyopathy. Matrix metalloproteinases (MMPs) are regulators of the ECM and can degrade all the components in the matrix. METHODS: Adolescent male mice were infected with cardiovirulent CVB3 and sacrificed at 3, 9, and 30 days post infection (pi). Transcription of MMP-2, MMP-9, and MMP-12 was assessed by reverse-transcriptase polymerase chain reaction (RT-PCR). Protein expression of these enzymes was examined using immunohistochemistry, and the activation status of MMP-2 and MMP-9 was assessed using gelatin zymography. Tissue inhibitors of metalloproteinases (TIMPs) were analyzed using immunoblotting assays. Myocarditic hearts were also stained with picrosirius red and viewed under polarizing light to examine the collagen network. RESULTS: MMP-2, MMP-9, and MMP-12 transcription was increased at 9 days pi, as determined by RT-PCR. Immunohistochemistry confirmed an increase in translation of these MMP species, and zymographic analysis further showed elevated activation of MMP-2 and MMP-9 following CVB3 infection. TIMP-3 and TIMP-4 expression was down-regulated, while TIMP-1 and TIMP-2 remained constant throughout the infection. Mouse hearts stained with picrosirius red showed an increase in total amount of collagen during the acute phase of infection and disrupted fibrils at later timepoints. CONCLUSION: After CVB3 infection, ECM remodeling is triggered, and this response may involve increased expression and activation of MMPs.


Subject(s)
Coxsackievirus Infections/physiopathology , Matrix Metalloproteinases/physiology , Myocarditis/physiopathology , Tissue Inhibitor of Metalloproteinases/physiology , Animals , Coxsackievirus Infections/enzymology , Down-Regulation , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , In Situ Hybridization , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myocarditis/enzymology , Myocarditis/virology , Myocardium/pathology , Up-Regulation
14.
Methods Mol Med ; 112: 109-54, 2005.
Article in English | MEDLINE | ID: mdl-16010014

ABSTRACT

The use of specialized reporter genes to monitor real-time, tissue-specific transgene expression in animal models offers an opportunity to circumvent current limitations associated with the establishment of transgenic mouse models. The Cre-loxP and the tetracycline (Tet)-inducible systems are useful methods of conditional gene expression that allow spatial (cell-type-specific) and temporal (inducer-dependent) control. Most often, the alpha-myosin heavy chain (alpha-MHC) promoter is used in these inducible systems to restrict expression of reporter genes and transgenes to the myocardium. An overview of each inducible system is described, along with suggested reporter genes for real-time, noninvasive imaging in the myocardium. Effective gene delivery of the inducible gene expression system is carried out by lentiviral vectors, which offer high transduction efficiency, long-term transgene expression, and low immunogenicity. This chapter outlines the packaging of myocardium-specific inducible expression systems into lentiviral vectors, in which a transgene and a reporter gene are transduced into cardiomyocytes. In doing so, transgene and reporter expression can be monitored/tracked with bioluminescence imaging (BLI) and positron emission tomography (PET).


Subject(s)
Gene Expression Regulation , Gene Transfer Techniques , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Repressor Proteins/genetics , Animals , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/metabolism , Image Processing, Computer-Assisted , Integrases/genetics , Lac Operon , Lentivirus/genetics , Luciferases/metabolism , Myocardium/cytology , Positron-Emission Tomography , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Recombination, Genetic , Repressor Proteins/metabolism , Tetracycline/pharmacology , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Transcriptional Activation , Transgenes , Viral Proteins/genetics
15.
Am J Physiol Endocrinol Metab ; 282(5): E1039-45, 2002 May.
Article in English | MEDLINE | ID: mdl-11934668

ABSTRACT

Glycolysis, measured by (3)H(2)O production from [5-(3)H]glucose, is accelerated in isolated working hypertrophied rat hearts. However, nonglycolytic detritiation of [5-(3)H]glucose via the nonoxidative pentose phosphate pathway (PPP) could potentially lead to an overestimation of true glycolytic rates, especially in hypertrophied hearts where the PPP may be upregulated. To address this concern, we measured glycolysis using [5-(3)H]glucose and a second, independent method in isolated working hearts from halothane-anesthetized, sham-operated and aortic-constricted rats. Glycolysis was accelerated in hypertrophied hearts compared with control hearts regardless of the method used. There was also excellent concordance in glycolytic rates between the different methods. Moreover, activity of glucose-6-phosphate dehydrogenase and expression of transaldolase, enzymes controlling key steps in the oxidative and nonoxidative PPP, respectively, were not different between control and hypertrophied hearts. Thus nonglycolytic detritiation of [5-(3)H]glucose in the PPP is insignificant, and (3)H(2)O production from [5-(3)H]glucose is an accurate means to measure glycolysis in isolated working normal and hypertrophied rat hearts. Furthermore, the PPP does not appear to be increased in cardiac hypertrophy induced by abdominal aortic constriction.


Subject(s)
Artifacts , Glycolysis/physiology , Hypertrophy, Left Ventricular/metabolism , Animals , Blood Pressure , Glucose/pharmacokinetics , Glucosephosphate Dehydrogenase/metabolism , Heart Rate , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Lactic Acid/metabolism , Male , Myocardium/enzymology , Myocardium/pathology , Organ Size , Oxidation-Reduction , Pentose Phosphate Pathway/physiology , Pyruvic Acid/metabolism , Rats , Rats, Sprague-Dawley , Transaldolase/metabolism , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...