Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nat Immunol ; 21(10): 1219-1231, 2020 10.
Article in English | MEDLINE | ID: mdl-32778760

ABSTRACT

Chronic inflammation is a common feature of obesity, with elevated cytokines such as interleukin-1 (IL-1) in the circulation and tissues. Here, we report an unconventional IL-1R-MyD88-IRAK2-PHB/OPA1 signaling axis that reprograms mitochondrial metabolism in adipocytes to exacerbate obesity. IL-1 induced recruitment of IRAK2 Myddosome to mitochondria outer membranes via recognition by TOM20, followed by TIMM50-guided translocation of IRAK2 into mitochondria inner membranes, to suppress oxidative phosphorylation and fatty acid oxidation, thereby attenuating energy expenditure. Adipocyte-specific MyD88 or IRAK2 deficiency reduced high-fat-diet-induced weight gain, increased energy expenditure and ameliorated insulin resistance, associated with a smaller adipocyte size and increased cristae formation. IRAK2 kinase inactivation also reduced high-fat diet-induced metabolic diseases. Mechanistically, IRAK2 suppressed respiratory super-complex formation via interaction with PHB1 and OPA1 upon stimulation of IL-1. Taken together, our results suggest that the IRAK2 Myddosome functions as a critical link between inflammation and metabolism, representing a novel therapeutic target for patients with obesity.


Subject(s)
Adipocytes/immunology , Inflammation/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1/metabolism , Mitochondrial Membranes/metabolism , Obesity/immunology , Adipocytes/pathology , Animals , Cells, Cultured , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Oxidative Phosphorylation , Prohibitins , Protein Transport , Receptors, Interleukin-1/metabolism , Signal Transduction
2.
J Reconstr Microsurg ; 34(1): 41-46, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29032571

ABSTRACT

BACKGROUND: The purpose of this study was to: (1) evaluate the mechanism of lymph drainage through a vascularized lymph node (VLN) flap, and (2) investigate if the number of VLNs impacts lymph transit time through the flap. METHODS: Twenty-seven axillary VLN flaps were elevated in 14 Sprague-Dawley rats and divided into three groups (n = 9 each) based on the number of lymph nodes present: group 1 (0-VLNs), group 2 (2-VLNs), and group 3 (4-VLNs). Indocyanine green (n = 8/group) and Alexa680-albumin (n = 1/group) were injected into the edge of flaps and the latency period between injection and fluorescence in the axillary vein was recorded. Stereomicroscopic fluorescent lymphography was performed to directly visualize lymphatic transit through VLNs. RESULTS: Fluorescence was detected in the axillary vein after 229s [47-476], 79s [15-289], and 56s [16-110] in group 1, 2, and 3, respectively (p < 0.01). There was a negative correlation between the number of VLNs in the flap and the latency period (r = -0.59; p < 0.05). Median flap weights were comparable in group 1, 2, and 3 (258 mg [196-349], 294 mg [212-407], 315 mg [204-386], respectively; p = 0.54). Stereoscopic lymphography allowed direct visualization of lymphatic fluid transit through VLNs. CONCLUSION: Lymphatic fluid in VLN flaps drains into the venous system mainly by passing through the afferent lymphatics and lymph nodes. A secondary mechanism appears to be the diffusion of fluid into the venous system via intratissue lymphaticovenous connections created during flap elevation. Increasing the number of lymph nodes in the flap is associated with a more rapid transit of fluid.


Subject(s)
Axilla/surgery , Lymph Nodes/transplantation , Lymphatic System/physiology , Surgical Flaps/blood supply , Animals , Coloring Agents , Disease Models, Animal , Drainage , Indocyanine Green , Lymph Nodes/blood supply , Lymph Nodes/innervation , Lymphography , Male , Rats , Rats, Sprague-Dawley
3.
Mol Cancer ; 13: 70, 2014 Mar 23.
Article in English | MEDLINE | ID: mdl-24655592

ABSTRACT

BACKGROUND: Macroautophagy is a catabolic process that can mediate cell death or survival. Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment (TR) is known to induce autophagy. Here we investigated whether SQSTM1/p62 (p62) overexpression, as a marker of autophagic flux, was related to aggressiveness of human prostate cancer (PCa) and whether autophagy regulated the treatment response in sensitive but not resistant PCa cell lines. METHODS: Immunostaining and immunoblotting analyses of the autophagic markers p62 [in PCa tissue microarrays (TMAs) and PCa cell lines] and LC3 (in PCa cell lines), transmission electron microscopy, and GFP-mCherry-LC3 were used to study autophagy induction and flux. The effect of autophagy inhibition using pharmacologic (3-methyladenine and chloroquine) and genetic [(short hairpin (sh)-mediated knock-down of ATG7 and LAMP2) and small interfering (si)RNA-mediated BECN1 knock-down] approaches on TR-induced cell death was assessed by clonogenic survival, sub-G1 DNA content, and annexinV/PI staining by flow cytometry. Caspase-8 activation was determined by immunoblotting. RESULTS: We found that increased cytoplasmic expression of p62 was associated with high-grade PCa, indicating that autophagy signaling might be important for survival in high-grade tumors. TR-resistant cells exhibited high autophagic flux, with more efficient clearance of p62-aggregates in four TR-resistant PCa cell lines: C4-2, LNCaP, DU145, and CWRv22.1. In contrast, autophagic flux was low in TR-sensitive PC3 cells, leading to accumulation of p62-aggregates. Pharmacologic (chloroquine or 3-methyladenine) and genetic (shATG7 or shLAMP2) inhibition of autophagy led to cell death in TR-resistant C4-2 cells. shATG7-expressing PC3 cells, were less sensitive to TR-induced cell death whereas those shLAMP2-expressing were as sensitive as shControl-expressing PC3 cells. Inhibition of autophagic flux using chloroquine prevented clearance of p62 aggregates, leading to caspase-8 activation and cell death in C4-2 cells. In PC3 cells, inhibition of autophagy induction prevented p62 accumulation and hence caspase-8 activation. CONCLUSIONS: We show that p62 overexpression correlates with advanced stage human PCa. Pharmacologic and genetic inhibition of autophagy in PCa cell lines indicate that autophagic flux can determine the cellular response to TR by regulating caspase-8 activation. Thus, combining various autophagic inhibitors may have a differential impact on TR-induced cell death.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Antineoplastic Agents/pharmacology , Autophagy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/physiology , Drug Resistance, Neoplasm/physiology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Sequestosome-1 Protein , Tissue Array Analysis
4.
J Biol Chem ; 286(30): 26849-59, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21636573

ABSTRACT

RIL (product of PDLIM4 gene) is an actin-associated protein that has previously been shown to stimulate actin bundling by interacting with actin-cross-linking protein α-actinin-1 and increasing its affinity to filamentous actin. Here, we report that the alternatively spliced isoform of RIL, denoted here as RILaltCterm, functions as a dominant-negative modulator of RIL-mediated actin reorganization. RILaltCterm is regulated at the level of protein stability, and this protein isoform accumulates particularly in response to oxidative stress. We show that the alternative C-terminal segment of RILaltCterm has a disordered structure that directs the protein to rapid degradation in the core 20 S proteasomes. Such degradation is ubiquitin-independent and can be blocked by binding to NAD(P)H quinone oxidoreductase NQO1, a detoxifying enzyme induced by prolonged exposure to oxidative stress. We show that either overexpression of RILaltCterm or its stabilization by stresses counteracts the effects produced by full-length RIL on organization of actin cytoskeleton and cell motility. Taken together, the data suggest a mechanism for fine-tuning actin cytoskeleton rearrangement in response to stresses.


Subject(s)
Actins/metabolism , Alternative Splicing , Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Oxidative Stress , Actinin/genetics , Actinin/metabolism , Actins/genetics , Animals , Cytoskeleton/genetics , DNA-Binding Proteins/genetics , Dogs , HeLa Cells , Humans , LIM Domain Proteins , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
Pathogens ; 10(4)2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33916455

ABSTRACT

In this study, we investigated stage specific expression, trafficking, solubility and topology of endogenous PfMC-2TM in P. falciparum (3D7) infected erythrocytes. Following Brefeldin A (BFA) treatment of parasites, PfMC-2TM traffic was evaluated using immunofluorescence with antibodies reactive with PfMC-2TM. PfMC-2TM is sensitive to BFA treatment and permeabilization of infected erythrocytes with streptolysin O (SLO) and saponin, showed that the N and C-termini of PfMC-2TM are exposed to the erythrocyte cytoplasm with the central portion of the protein protected in the MC membranes. PfMC-2TM was expressed as early as 4 h post invasion (hpi), was tightly colocalized with REX-1 and trafficked to the erythrocyte membrane without a change in solubility. PfMC-2TM associated with the MC and infected erythrocyte membrane and was resistant to extraction with alkaline sodium carbonate, suggestive of protein-lipid interactions with membranes of the MC and erythrocyte. PfMC-2TM is an additional marker of the nascent MCs.

6.
J Assist Reprod Genet ; 27(2-3): 97-101, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20140639

ABSTRACT

OBJECTIVE: A simple method for preparation of isolated ovarian follicles for transmission electron microscopy (TEM) using transwell inserts is described. MATERIALS AND METHODS: Pre-antral follicles were enzymatically isolated from mouse ovaries and cultured overnight on transwell insert polyester membranes. The following day, isolated ovarian follicles were processed for TEM by moving the transwell insert through successive wells containing the fixation and embedding reagents. After polymerization of the resin, the polyester membrane with the follicles embedded in the resin was disengaged from the transwell unit. The resin was sectioned. Semi-thin sections were stained with toluidine blue while ultra-thin sections were stained by uranyl acetate and examined by light microscopy and TEM, respectively. RESULTS: Isolated ovarian follicles were easily processed in groups for TEM. Follicles were well embedded and there appeared to be no loss of tissue during processing. The ultra-structure of processed isolated ovarian follicles was well preserved with little evidence of processing artifacts. CONCLUSIONS: In situ processing and preparation of isolated ovarian follicles by first allowing attachment on transwell insert membranes was shown to be a simple, rapid and effective method for TEM.


Subject(s)
Microscopy, Electron , Ovarian Follicle/ultrastructure , Animals , Cell Adhesion , Crosses, Genetic , Female , Membranes, Artificial , Mice , Mice, Inbred C57BL , Organ Culture Techniques/instrumentation , Organ Culture Techniques/methods , Polyesters , Specimen Handling , Staining and Labeling/methods , Tissue Embedding/methods , Tissue Fixation/methods
7.
Mol Biol Cell ; 17(2): 977-89, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16339081

ABSTRACT

Syntaxins 3 and 4 localize to the apical and basolateral plasma membrane, respectively, of epithelial cells where they mediate vesicle fusion. Here, we report that before establishment of cell polarity, syntaxins 3 and 4 are confined to mutually exclusive, submicron-sized clusters. Syntaxin clusters are remarkably uniform in size, independent of expression levels, and are distinct from caveolae and clathrin-coated pits. SNAP-23 partially colocalizes with both syntaxin 3 and 4 clusters. Deletion of the apical targeting signal of syntaxin 3 does not prevent sorting into clusters away from syntaxin 4. Syntaxin 3 and 4 cluster formation depends on different mechanisms because the integrity of syntaxin 3 clusters depends on intact microtubules, whereas syntaxin 4 clusters depend on intact actin filaments. Cholesterol depletion causes dispersion of syntaxin 3 but not syntaxin 4 clusters. In migrating cells, syntaxin clusters polarize to the leading edge, suggesting a role in polarized exocytosis. These results suggest that exocytosis occurs at small fusion sites exhibiting high local concentrations of SNARE proteins that may be required for efficient membrane fusion. The establishment of separate clusters for each syntaxin suggests that the plasma membrane is inherently polarized on an ultrastructural level even before the establishment of true cell polarity.


Subject(s)
Cell Membrane/chemistry , Cell Polarity , Qa-SNARE Proteins/analysis , Actins/physiology , Actins/ultrastructure , Animals , Caveolae/ultrastructure , Cell Line , Cell Membrane/ultrastructure , Cholesterol/metabolism , Coated Pits, Cell-Membrane/ultrastructure , Dogs , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Jurkat Cells , Mice , Microtubules/physiology , Microtubules/ultrastructure , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/analysis , Qc-SNARE Proteins/analysis , Sequence Deletion , Syntaxin 1/analysis
8.
Eur J Protistol ; 71: 125637, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31689662

ABSTRACT

In this study, we investigated morphological, immunological and molecular characteristics of Colpodella sp. (American Type Culture Collection 50594) in a diprotist culture containing Bodo caudatus as prey using Plasmodium rhoptry specific antibodies and oligonucleotide primers targeting Plasmodium falciparum rhoptry genes. In culture, Colpodella sp. attached to its prey using the apical end with attachment lasting for approximately 20 min while the cytoplasmic contents of the prey were aspirated into the posterior food vacuole of Colpodella sp. Encystment of Colpodella sp. was observed following feeding. Indirect immunofluorescence assay (IFA) and confocal microscopy using P. falciparum rhoptry specific antibodies showed intense reactivity with cytoplasmic vesicles of Colpodella sp. Bodo caudatus from diprotist and monoprotist (ATCC 30395) cultures showed weak background reactivity. Giemsa staining permitted differentiation of both protists. Genomic DNA isolated from the diprotist culture was used in polymerase chain reaction (PCR) with oligonucleotide primers targeting the P. falciparum rhoptry genes RhopH3, RhopH1/Clag3.2 and RAMA. Primers targeting exon 7 of the P. falciparum RhopH3 gene amplified an approximately 2 kb DNA fragment from the diprotist DNA template. DNA sequence and BLAST search analysis of the amplified product from diprotist DNA identified the RhopH3 gene demonstrating that the RhopH3 gene is conserved in Colpodella sp.


Subject(s)
Alveolata/genetics , Protozoan Proteins/genetics , Conserved Sequence
9.
J Biomed Mater Res A ; 107(11): 2522-2535, 2019 11.
Article in English | MEDLINE | ID: mdl-31334591

ABSTRACT

Targeted therapies provide increased efficiency for the detection and treatment of cancer with reduced side effects. Folate receptor (alpha subunit) is overexpressed in multiple tumors including liver cancer. In this study, we evaluated the specificity and toxicity of a folic acid-containing drug delivery vehicle (DDV) in a hepatocellular carcinoma (HCC) model. The DDV was prepared with two units each of folic acid (FA) and fluorescein isothiocyanate (FITC) molecules and conjugated to a central poly (ethylene glycol) (PEG) core via a modified chemo-enzymatic synthetic process. Rat hepatoma (N1S1) and human monocytic (U937) cell lines were used for cell culture-based assays and tested for DDV uptake and toxicity. Folate receptor expressions in liver tissues and cell lines were verified using standard immunohistochemistry techniques. Rat HCC model was used for in vivo assessment. The DDV was injected via intra-arterial or intravenous methods and imaged with IVIS spectrum in vivo imaging system. Strong signals of FITC in the liver tumor region correlated to targeted DDV uptake. The use of PEG enhanced water-solubility and provided flexibility for the interaction of FA ligands with multiple cell surface folate receptors that resulted in increased specific uptake. Our study suggested that PEG incorporation and folate targeting via intra-arterial approach is an efficient strategy for targeted delivery in HCC therapy.


Subject(s)
Carcinoma, Hepatocellular , Drug Delivery Systems , Fluorescein-5-isothiocyanate , Folic Acid , Liver Neoplasms, Experimental , Optical Imaging , Animals , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Rats , Rats, Sprague-Dawley , U937 Cells
10.
Exp Parasitol ; 120(1): 113-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606406

ABSTRACT

The biogenesis, organization and function of the rhoptries are not well understood. Antisera were prepared to synthetic peptides prepared as multiple antigenic peptides (MAPs) obtained from a Plasmodium yoelii merozoite rhoptry proteome analysis. The antisera were used in immunofluorescence and immunoelectron microscopy of schizont-infected erythrocytes. Twenty-seven novel rhoptry proteins representing proteases, metabolic enzymes, secreted proteins and hypothetical proteins, were identified in the body of the rhoptries by immunoelectron microscopy. The merozoite rhoptries contain a heterogeneous mixture of proteins that may initiate host cell invasion and establish intracellular parasite development.


Subject(s)
Malaria/parasitology , Plasmodium yoelii/chemistry , Protozoan Proteins/analysis , Animals , Female , Fluorescent Antibody Technique, Indirect , Male , Merozoites/chemistry , Merozoites/ultrastructure , Mice , Microscopy, Immunoelectron , Organelles/chemistry , Organelles/ultrastructure , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Plasmodium yoelii/ultrastructure , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Rabbits
11.
Cell Rep ; 23(2): 485-498, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642006

ABSTRACT

Focal adhesions anchor cells to extracellular matrix (ECM) and direct assembly of a pre-stressed actin cytoskeleton. They act as a cellular sensor and regulator, linking ECM to the nucleus. Here, we identify proteolytic turnover of the anti-adhesive proteoglycan versican as a requirement for maintenance of smooth muscle cell (SMC) focal adhesions. Using conditional deletion in mice, we show that ADAMTS9, a secreted metalloprotease, is required for myometrial activation during late gestation and for parturition. Through knockdown of ADAMTS9 in uterine SMC, and manipulation of pericellular versican via knockdown or proteolysis, we demonstrate that regulated pericellular matrix dynamics is essential for focal adhesion maintenance. By influencing focal adhesion formation, pericellular versican acts upstream of cytoskeletal assembly and SMC differentiation. Thus, pericellular versican proteolysis by ADAMTS9 balances pro- and anti-adhesive forces to maintain an SMC phenotype, providing a concrete example of the dynamic reciprocity of cells and their ECM.


Subject(s)
ADAMTS9 Protein/metabolism , Focal Adhesions/metabolism , ADAMTS9 Protein/antagonists & inhibitors , ADAMTS9 Protein/genetics , Actins/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Female , Humans , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Myometrium/pathology , Pregnancy , RNA Interference , RNA, Small Interfering/metabolism , Uterus/cytology , Versicans/metabolism
12.
Mol Cell Biol ; 22(7): 2398-409, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884622

ABSTRACT

Cyclin E/Cdk2 is a critical regulator of cell cycle progression from G(1) to S in mammalian cells and has an established role in oncogenesis. Here we examined the role of deregulated cyclin E expression in apoptosis. The levels of p50-cyclin E initially increased, and this was followed by a decrease starting at 8 h after treatment with genotoxic stress agents, such as ionizing radiation. This pattern was mirrored by the cyclin E-Cdk2-associated kinase activity and a time-dependent expression of a novel p18-cyclin E. p18-cyclin E was induced during apoptosis triggered by multiple genotoxic stress agents in all hematopoietic tumor cell lines we have examined. The p18-cyclin E expression was prevented by Bcl-2 overexpression and by the general caspase and specific caspase 3 pharmacologic inhibitors zVAD-fluoromethyl ketone (zVAD-fmk) and N-acetyl-Asp-Glu-Val-Asp-aldehyde (DEVD-CHO), indicating that it was linked to apoptosis. A p18-cyclin E(276-395) (where cyclin E(276-395) is the cyclin E fragment containing residues 276 to 395) was reconstituted in vitro, with mutagenesis experiments, indicating that the caspase-dependent cleavage was at amino acid residues 272 to 275. Immunoprecipitation analyses of the ectopically expressed cyclin E(1-275), cyclin E(276-395) deletion mutants, and native p50-cyclin E demonstrated that caspase-mediated cyclin E cleavage eliminated interaction with Cdk2 and therefore inactivated the associated kinase activity. Overexpression of cyclin E(276-395), but not of several other cyclin E mutants, specifically induced phosphatidylserine exposure and caspase activation in a dose-dependent manner, which were inhibited in Bcl-2-overexpressing cells or in the presence of zVAD-fmk. Apoptosis and generation of p18-cyclin E were significantly inhibited by overexpressing the cleavage-resistant cyclin E mutant, indicating a functional role for caspase-dependent proteolysis of cyclin E for apoptosis of hematopoietic tumor cells.


Subject(s)
Apoptosis , CDC2-CDC28 Kinases , Cyclin E/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Protein Processing, Post-Translational , Apoptosis/radiation effects , Blotting, Western , Caspases/metabolism , Cyclin E/chemistry , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , DNA Damage/radiation effects , Dose-Response Relationship, Radiation , Flow Cytometry , Hematopoietic Stem Cells/radiation effects , Humans , Molecular Weight , Mutagenesis, Site-Directed , Protein Processing, Post-Translational/radiation effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins , Tumor Cells, Cultured
13.
Laryngoscope ; 117(9): 1615-21, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17667133

ABSTRACT

OBJECTIVES/HYPOTHESIS: Dendritic cells (DC) are potent antigen-presenting cells that instigate allograft rejection. Their migration kinetics vary depending on the type of organ transplanted. The timing of donor and recipient DC trafficking in laryngeal transplants is unknown. STUDY DESIGN: Prospective animal model. METHODS: Lewis to Brown Norway (BN) rat laryngeal allografts and BN to BN isografts were performed without immunosuppression. Recipient animals were sacrificed at seven posttransplant time points. Total DC, as well as recipient and donor DC (in allograft recipients), were enumerated in situ in the airway epithelium and subepithelium using monoclonal antibodies, immunofluorescence, confocal microscopy, and image analysis software. RESULTS: Total DC densities in both laryngeal allografts and isografts decreased to approximately 10% of their initial values in the first 3 days and then rose beyond their starting values. In allografts, there was a net efflux of donor DC, reaching a nadir by 3 to 5 days; they were identified in recipient cervical lymph nodes from 12 hours to 5 days. Recipient DC infiltrated the laryngeal allograft, reaching a maximal density by day 7. CONCLUSION: The paradigm of donor DC efflux and recipient DC influx has been confirmed in a rat laryngeal transplant model, and the allograft-specific timing of these events has been elucidated. Similarities in total DC migration between allografts and isografts suggest that this phenomenon may not be driven entirely by major histocompatibility mismatch. Further understanding of trafficking may help with the goal of manipulating DC to induce allograft tolerance in the absence of generalized immunosuppression.


Subject(s)
Cell Movement/physiology , Dendritic Cells/physiology , Larynx/transplantation , Animals , Antibodies, Monoclonal , Antigens, Differentiation/metabolism , Fluorescent Antibody Technique , Larynx/immunology , Male , Prospective Studies , Rats , Rats, Inbred Lew , Tissue Donors , Transplantation
14.
Ann Otol Rhinol Laryngol ; 116(6): 449-56, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17672248

ABSTRACT

OBJECTIVES: Dendritic cells (DCs) are key instigators of rejection after transplantation. Their distribution has not been systematically characterized in all locations of the larynx and its surrounding tissues. METHODS: Rat larynges were stained with monoclonal antibodies identifying DCs. These cells were then enumerated by a new combination of techniques including immunofluorescence, confocal microscopy, and imaging software. RESULTS: The vast majority of DCs were located in the epithelium and subepithelium of the airway; the mean DC density ranged from 9 cells per square millimeter (0.2% of cells) to 645 cells per square millimeter (10.3% of cells). Their density in the epithelium was 3 to 11 times higher than that in the subepithelium. Non-airway sites (thyroid, parathyroid, muscle, fat) had mean DC densities varying from 3 cells per square millimeter (0.2%) to 57 cells per square millimeter (0.8%). No DCs were detected in cartilage. CONCLUSIONS: Dendritic cells are concentrated in the laryngotracheal epithelium and subepithelium and represent a much smaller proportion in the other sites studied. A baseline for laryngeal DC population studies has been established, and a computerized model for consistent quantitation using confocal microscopy has been developed. This unique method will serve as a foundation for investigating DC trafficking after rat laryngeal transplantation.


Subject(s)
Antigens, Differentiation/metabolism , Dendritic Cells/metabolism , Larynx/cytology , Larynx/metabolism , Animals , Fluorescent Antibody Technique , Male , Rats , Rats, Inbred Lew , Trachea/cytology , Trachea/metabolism
15.
Acta Biomater ; 60: 210-219, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28709984

ABSTRACT

Current clinical methods to treat articular cartilage lesions provide temporary relief of the symptoms but fail to permanently restore the damaged tissue. Tissue engineering, using mesenchymal stem cells (MSCs) combined with scaffolds and bioactive factors, is viewed as a promising method for repairing cartilage injuries. However, current tissue engineered constructs display inferior mechanical properties compared to native articular cartilage, which could be attributed to the lack of structural organization of the extracellular matrix (ECM) of these engineered constructs in comparison to the highly oriented structure of articular cartilage ECM. We previously showed that we can guide MSCs undergoing chondrogenesis to align using microscale guidance channels on the surface of a two-dimensional (2-D) collagen scaffold, which resulted in the deposition of aligned ECM within the channels and enhanced mechanical properties of the constructs. In this study, we developed a technique to roll 2-D collagen scaffolds containing MSCs within guidance channels in order to produce a large-scale, three-dimensional (3-D) tissue engineered cartilage constructs with enhanced mechanical properties compared to current constructs. After rolling the MSC-scaffold constructs into a 3-D cylindrical structure, the constructs were cultured for 21days under chondrogenic culture conditions. The microstructure architecture and mechanical properties of the constructs were evaluated using imaging and compressive testing. Histology and immunohistochemistry of the constructs showed extensive glycosaminoglycan (GAG) and collagen type II deposition. Second harmonic generation imaging and Picrosirius red staining indicated alignment of neo-collagen fibers within the guidance channels of the constructs. Mechanical testing indicated that constructs containing the guidance channels displayed enhanced compressive properties compared to control constructs without these channels. In conclusion, using a novel roll-up method, we have developed large scale MSC based tissue-engineered cartilage that shows microscale structural organization and enhanced compressive properties compared to current tissue engineered constructs. STATEMENT OF SIGNIFICANCE: Tissue engineered cartilage constructs made with human mesenchymal stem cells (hMSCs), scaffolds and bioactive factors are a promising solution to treat cartilage defects. A major disadvantage of these constructs is their inferior mechanical properties compared to the native tissue, which is likely due to the lack of structural organization of the extracellular matrix of the engineered constructs. In this study, we developed three-dimensional (3-D) cartilage constructs from rectangular scaffold sheets containing hMSCs in micro-guidance channels and characterized their mechanical properties and metabolic requirements. The work led to a novel roll-up method to embed 2-D microscale structures in 3-D constructs. Further, micro-guidance channels incorporated within the 3-D cartilage constructs led to the production of aligned cell-produced matrix and enhanced mechanical function.


Subject(s)
Cartilage/metabolism , Chondrogenesis , Collagen/chemistry , Mesenchymal Stem Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cartilage/cytology , Cattle , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology
16.
Invest Ophthalmol Vis Sci ; 47(1): 241-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16384969

ABSTRACT

PURPOSE: The infiltration of inflammatory cells into the cornea is a major determinant in the outcome of keratitis. The purpose of this study was to use enhanced green fluorescence protein (EGFP) bone marrow chimeric mice to visualize and characterize the inflammatory cells that migrate to the corneal stroma during endotoxin-induced keratitis and to explore the mechanisms underlying this process. METHODS: Keratitis was induced by injecting endotoxin into the corneas of EGFP chimeric mice. In vivo fluorescence stereomicroscopy was used to visualize in real time the recruitment of EGFP-positive cells at different time points. Immunohistochemistry and three-dimensional (3D) confocal analysis of whole-mount corneas was used for histologic characterization. Macrophage inflammatory protein-2 (MIP-2) chemokine was neutralized in vivo to determine its contribution to this process. RESULTS: Recruitment of EGFP-positive inflammatory cells in the corneal stroma can be detected in vivo by 6 hours after the injection of endotoxin, and these were mainly neutrophils. Full-thickness whole corneal mount confocal image analysis showed a distinct pattern of migration of EGFP inflammatory cells through the anterior corneal stroma. Moreover, inflammatory cells did not colocalize with the injected lipopolysaccharide (LPS) deposits in the stroma but moved from all directions toward LPS, partially in response to the production of the chemokine MIP-2. CONCLUSIONS: EGFP chimeric mice and ex vivo 3D analysis of whole-mount corneas provides unique information on the interaction of infiltrating inflammatory cells in the cornea. These findings demonstrate that a chemotactic gradient triggered in part by MIP-2 is responsible for directing inflammatory cell migration through the corneal stroma.


Subject(s)
Bone Marrow Cells/physiology , Chemotaxis, Leukocyte/physiology , Corneal Stroma/immunology , Keratitis/immunology , Animals , Cell Movement/physiology , Chemokine CXCL2 , Chemokines/metabolism , Corneal Stroma/pathology , Green Fluorescent Proteins/metabolism , Keratitis/chemically induced , Keratitis/pathology , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Neutrophils/physiology , Transplantation Chimera
17.
Sarcoidosis Vasc Diffuse Lung Dis ; 23(2): 93-100, 2006 Jun.
Article in English | MEDLINE | ID: mdl-17937104

ABSTRACT

BACKGROUND AND AIM: Recent evidence suggests that the transcription factor, PPARgamma, is an important negative regulator of inflammation. Because studies of murine adipocytes and macrophages implicate IFN-gamma, a key mediator of granuloma formation in sarcoidosis, as a PPARgamma antagonist, we investigated the relationship between PPARgamma and IFN-gamma in bronchoalveolar lavage (BAL) cells of sarcoidosis patients and healthy controls. METHODS: BAL cells were analyzed for PPARgamma and IFN-gamma mRNA expression by quantitative PCR and for PPARgamma protein by immunocytochemistry and western blotting. RESULTS: In sarcoidosis patients with severe, treatment-requiring disease, IFN-gamma was strikingly elevated and PPARgamma gene expression was deficient. In contrast, PPARgamma expression of non-severe patients was comparable to control but was still accompanied by increased IFN-gamma. By confocal microscopy, nuclear PPARgamma protein was detectable in alveolar macrophages from non-severe patients unlike previous observations of severe patients. In vitro exposure of BAL cells or purified alveolar macrophages to IFN-gamma resulted in dose-dependent repression of PPARgamma mRNA in both sarcoidosis and controls. IFN-gamma treatment also reduced PPARgamma protein in BAL lysates and nuclear PPARgamma content in control alveolar macrophages, resulting in a diffuse cytoplasmic PPARgamma distribution similar to that observed in severe sarcoidosis. CONCLUSION: These novel results indicate that IFN-gamma represses PPARgamma in human alveolar macrophages but that in sarcoidosis, PPARgamma rather than IFN-gamma levels correlate best with disease severity. Data also emphasize the complex nature of PPARgamma restorative mechanisms in alveolar macrophages exposed to an inflammatory environment containing IFN-gamma -- a potential PPARgamma antagonist.


Subject(s)
Gene Expression , Interferon-gamma/metabolism , PPAR gamma/metabolism , RNA, Messenger/genetics , Sarcoidosis, Pulmonary/metabolism , Biomarkers/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Female , Humans , Immunohistochemistry , Interferon-gamma/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Microscopy, Confocal , Middle Aged , PPAR gamma/genetics , Polymerase Chain Reaction , Prognosis , Sarcoidosis, Pulmonary/genetics , Sarcoidosis, Pulmonary/pathology , Severity of Illness Index
18.
J Natl Cancer Inst ; 94(13): 1010-9, 2002 Jul 03.
Article in English | MEDLINE | ID: mdl-12096086

ABSTRACT

BACKGROUND: The ubiquitous plasma membrane transcobalamin II receptor (TC II-R) mediates uptake of cobalamin (Cbl; vitamin B12), an essential micronutrient. Tumors often require more Cbl than normal tissue, and increased Cbl uptake may result from increased TC II-R expression. To examine whether Cbl could therefore be used as a carrier molecule to target a chemotherapy drug, we tested an analogue of Cbl with nitric oxide as a ligand, nitrosylcobalamin (NO-Cbl). Because interferon beta (IFN-beta) has antitumor effects and increases expression of some membrane receptors, we examined whether it may enhance the effects of NO-Cbl. METHODS: Antiproliferative effects of NO-Cbl were assessed in 24 normal and cancer cell lines. Xenograft tumors of human ovarian cancer NIH-OVCAR-3 cells were established in athymic nude mice, and tumor growth was monitored after treatment with NO-Cbl and IFN-beta, both individually and concomitantly. TC II-R expression and apoptosis was monitored in vitro and in vivo. RNA protection assays and mitochondrial membrane potential assays were used to distinguish the extrinsic and intrinsic apoptotic pathways, respectively. RESULTS: Cancer cell lines were more sensitive to NO-Cbl (with ID(50)s [the dose that inhibits growth by 50%] as low as 2 microM) than normal cell lines (with ID(50)s of 85-135 microM). Single-agent NO-Cbl and IFN-beta treatment of NIH-OVCAR-3 xenografts induced tumor regression, whereas combination treatment induced tumor eradication. IFN-beta treatment increased TC II-R expression in vitro and uptake of [(57)Co]cobalamin in vivo. Compared with NIH-OVCAR-3 cells treated with NO-Cbl, cells treated with NO-Cbl and IFN-beta were more apoptotic and expressed higher mRNA levels of various apoptosis-associated genes. No changes in mitochondrial membrane potential were observed in cells treated with NO-Cbl. CONCLUSION: NO-Cbl inhibited tumor growth in vivo by activating the extrinsic apoptotic pathway. The increased expression of TC II-R induced by IFN-beta resulted in enhanced antitumor effects with NO-Cbl both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , Interferon-beta/therapeutic use , Melanoma/therapy , Nitroso Compounds/pharmacology , Ovarian Neoplasms/therapy , Receptors, Cell Surface/metabolism , Vitamin B 12/analogs & derivatives , Vitamin B 12/pharmacology , Animals , Annexin A5/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Division/drug effects , Combined Modality Therapy , Drug Synergism , Female , Humans , Immunoenzyme Techniques , Male , Melanoma/metabolism , Melanoma/pathology , Membrane Potentials , Mice , Mice, Nude , Mitochondria/metabolism , Nitric Oxide/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rhodamines , Ribonuclease, Pancreatic/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
19.
Biochim Biophys Acta ; 1673(1-2): 3-12, 2004 Jul 06.
Article in English | MEDLINE | ID: mdl-15238245

ABSTRACT

A variety of obstacles have hindered the ultrastructural localization of hyaluronan (HA). These include a lack of adequate fixation techniques to prevent the loss of HA, the lack of highly sensitive and specific probes, and a lack of accessibility due to the masking of HA by HA-binding macromolecules such as proteoglycans and glycoproteins. Despite these problems, a number of studies, both biochemical and histochemical, have been published indicating that HA is not restricted to the extracellular milieu, but is also present intracellularly. This review focuses on the possible functions of intracellular HA, its potential relationships to extracellular HA structures, and implications for inflammatory processes.


Subject(s)
Hyaluronic Acid/metabolism , Inflammation/metabolism , Muscle, Smooth/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cells, Cultured , Colitis/metabolism , Cycloheximide , Diabetic Nephropathies/metabolism , Glucuronosyltransferase , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/chemistry , Hyaluronan Synthases , Hyaluronic Acid/analysis , Hyaluronic Acid/biosynthesis , Hyaluronic Acid/chemistry , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Microscopy, Confocal , Mitosis , Monocytes/immunology , Monocytes/metabolism , Muscle Cells/metabolism , Muscle, Smooth/cytology , Poly I-C , Transferases/metabolism
20.
Invest Ophthalmol Vis Sci ; 45(7): 2194-200, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15223795

ABSTRACT

PURPOSE: This study was conducted to determine whether intrastromal injection of adenoviral construct could be used to transfect corneal stroma cells effectively in vivo and to determine whether a tissue-specific promoter could be used to express exogenous genes in keratocytes. METHODS: An adenoviral construct with a cytomegalovirus (pCMV)-driven enhanced green fluorescent protein (EGFP) reporter gene was injected into the stroma of murine corneas. In vivo expression was quantitated and samples were analyzed by in vivo stereomicroscopy, and ex vivo expression was determined by confocal three dimensional (3-D) reconstruction. The 3.2-kb keratocan promoter was used to drive tissue-specific reporter gene expression in vivo. RESULTS: EGFP expression was first detected in vivo 11 hours after injection of adeno-EGFP in the corneal stroma, with a duration of approximately 3 weeks. Ex vivo wholemount cornea confocal analysis with 3-D reconstruction allowed visualization of EGFP expression in corneal stroma cells, to accurately assess cellular architecture and distribution in the corneal stroma. Naked pCMV-EGFP plasmid DNA did not express the reporter gene to the levels of the adeno-EGFP. The 3.2-kb keratocan promoter was capable of driving EGFP tissue-specific expression in the cornea. CONCLUSIONS: Intrastromal injection of adenovirus packaged DNA constructs is a rapid and efficient way to deliver and express genes in the corneal stroma. Intrastromal injection is also capable of delivering tissue-specific promoter constructs to the corneal stroma for gene expression. Furthermore, 3-D reconstruction provides a powerful tool for enhanced visualization of the corneal stroma environment and cellular biology.


Subject(s)
Adenoviridae/genetics , Corneal Stroma/metabolism , Genetic Vectors , Luminescent Proteins/genetics , Promoter Regions, Genetic , Proteoglycans/genetics , Transfection/methods , Animals , Fluorescent Antibody Technique, Indirect , Gene Expression , Green Fluorescent Proteins , Imaging, Three-Dimensional , Injections , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL