Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Orthopedics ; 36(10): e1295-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24093707

ABSTRACT

The classic literature describes the transverse humeral ligament (THL) as a distinct anatomic structure with a role in biceps tendon stability; however, recent literature suggests that it is not a distinct anatomic structure. The purpose of this study was to evaluate the gross and microscopic anatomy of the THL, including a specific investigation of the histology of this ligament. Thirty frozen, embalmed cadaveric specimens were dissected to determine the gross anatomy of the THL. Seven specimens were evaluated histologically for the presence of mechanoreceptors and free nerve endings. Two tissue layers were identified in the area described as the THL. In the deep layer, fibers of the subscapularis tendon were found to span the bicipital groove with contributions from the coracohumeral ligament and the supraspinatus tendon. Superficial to this layer was a fibrous fascial covering consisting of distinct bands of tissue. Neurohistology staining revealed the presence of free nerve endings but no mechanoreceptors. This study's findings demonstrate that the THL is a distinct structure continuous with the rotator cuff tendons and the coracohumeral ligament. The finding of free nerve endings in the THL suggests a potential role as a shoulder pain generator.


Subject(s)
Ligaments/innervation , Shoulder Joint/anatomy & histology , Humans
2.
PLoS One ; 7(7): e42088, 2012.
Article in English | MEDLINE | ID: mdl-22848713

ABSTRACT

Demyelination and axonal degeneration are determinants of progressive neurological disability in patients with multiple sclerosis (MS). Cells resident within the central nervous system (CNS) are active participants in development, progression and subsequent control of autoimmune disease; however, their individual contributions are not well understood. Astrocytes, the most abundant CNS cell type, are highly sensitive to environmental cues and are implicated in both detrimental and protective outcomes during autoimmune demyelination. Experimental autoimmune encephalomyelitis (EAE) was induced in transgenic mice expressing signaling defective dominant-negative interferon gamma (IFN-γ) receptors on astrocytes to determine the influence of inflammation on astrocyte activity. Inhibition of IFN-γ signaling to astrocytes did not influence disease incidence, onset, initial progression of symptoms, blood brain barrier (BBB) integrity or the composition of the acute CNS inflammatory response. Nevertheless, increased demyelination at peak acute disease in the absence of IFN-γ signaling to astrocytes correlated with sustained clinical symptoms. Following peak disease, diminished clinical remission, increased mortality and sustained astrocyte activation within the gray matter demonstrate a critical role of IFN-γ signaling to astrocytes in neuroprotection. Diminished disease remission was associated with escalating demyelination, axonal degeneration and sustained inflammation. The CNS infiltrating leukocyte composition was not altered; however, decreased IL-10 and IL-27 correlated with sustained disease. These data indicate that astrocytes play a critical role in limiting CNS autoimmune disease dependent upon a neuroprotective signaling pathway mediated by engagement of IFN-γ receptors.


Subject(s)
Astrocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-gamma/metabolism , Signal Transduction , Animals , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Mice , Mice, Inbred C57BL , Myelin Sheath/physiology , Receptors, Interferon/metabolism , Interferon gamma Receptor
3.
Hip Int ; 22(1): 75-81, 2012.
Article in English | MEDLINE | ID: mdl-22344482

ABSTRACT

Hip arthroscopy remains a useful surgical intervention for labral injuries. The literature has predominantly focused on structural and vascular considerations of the hip joint, with few studies examining the neurohistology of the surrounding periarticular tissues. We mapped and identified the periarticular neural anatomy, to identify the presence of sensory nerve fibres and mechanoreceptors within the hip joint. Eight human cadaveric hips were dissected into a total of ten specimens per hip. Histological staining was used to identify neural structures taken from the superolateral, anterior, inferior, and posterior positions of the hip joint. The frozen sections were analyzed by light microscopy to calculate relative concentrations of mean neural fibres per high power field (mnf/hpf). Neural end organs were found in the hip capsule, acetabular labrum, ligamentum teres and transverse acetabular ligament. The highest levels of mechanoreceptors were found in the superolateral aspect of the hip capsule (9.6 mnf/hpf). The labrum showed highest levels of sensory fibres (3.4 mnf/hpf) and mechanoreceptors (4.3mnf/hpf) within the anterior zone. Sensory fibres and mechanoreceptors densely populate the acetabular labrum, capsule and transverse acetabular ligament. The anterior zone of the labrum contained the highest relative concentration of sensory fibres, specifically Ruffini corpuscles.


Subject(s)
Hip Joint/innervation , Mechanoreceptors/cytology , Peripheral Nerves/anatomy & histology , Sensory Receptor Cells/cytology , Acetabulum/anatomy & histology , Aged , Aged, 80 and over , Cadaver , Cartilage, Articular/innervation , Female , Humans , Joint Capsule/innervation , Male
4.
Gastroenterology ; 142(4): 918-927.e6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22178212

ABSTRACT

BACKGROUND & AIMS: Capillarization, characterized by loss of differentiation of liver sinusoidal endothelial cells (LSECs), precedes the onset of hepatic fibrosis. We investigated whether restoration of LSEC differentiation would normalize crosstalk with activated hepatic stellate cells (HSC) and thereby promote quiescence of HSC and regression of fibrosis. METHODS: Rat LSECs were cultured with inhibitors and/or agonists and examined by scanning electron microscopy for fenestrae in sieve plates. Cirrhosis was induced in rats using thioacetamide, followed by administration of BAY 60-2770, an activator of soluble guanylate cyclase (sGC). Fibrosis was assessed by Sirius red staining; expression of α-smooth muscle actin was measured by immunoblot analysis. RESULTS: Maintenance of LSEC differentiation requires vascular endothelial growth factor-A stimulation of nitric oxide-dependent signaling (via sGC and cyclic guanosine monophosphate) and nitric oxide-independent signaling. In rats with thioacetamide-induced cirrhosis, BAY 60-2770 accelerated the complete reversal of capillarization (restored differentiation of LSECs) without directly affecting activation of HSCs or fibrosis. Restoration of differentiation to LSECs led to quiescence of HSCs and regression of fibrosis in the absence of further exposure to BAY 60-2770. Activation of sGC with BAY 60-2770 prevented progression of cirrhosis, despite continued administration of thioacetamide. CONCLUSIONS: The state of LSEC differentiation plays a pivotal role in HSC activation and the fibrotic process.


Subject(s)
Capillaries/pathology , Cell Differentiation , Endothelial Cells/pathology , Hepatic Stellate Cells/pathology , Liver Cirrhosis, Experimental/pathology , Liver/blood supply , Liver/pathology , Paracrine Communication , Actins/metabolism , Animals , Benzoates/pharmacology , Biphenyl Compounds , Blotting, Western , Capillaries/drug effects , Capillaries/metabolism , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Cyclic GMP/metabolism , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation , Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hydrocarbons, Fluorinated/pharmacology , Liver/drug effects , Liver/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/prevention & control , Male , Microscopy, Electron, Scanning , Nitric Oxide , Paracrine Communication/drug effects , Phenotype , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Soluble Guanylyl Cyclase , Thioacetamide , Vascular Endothelial Growth Factor A/metabolism
5.
J Histochem Cytochem ; 59(12): 1087-100, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22034518

ABSTRACT

Progress in our understanding of heterotypic cellular interaction in the tumor microenvironment, which is recognized to play major roles in cancer progression, has been hampered due to unavailability of an appropriate in vitro co-culture model. The aim of this study was to generate an in vitro 3-dimensional human breast cancer model, which consists of cancer cells and fibroblasts. Breast cancer cells (UACC-893) and fibroblasts at various densities were co-cultured in a rotating suspension culture system to establish co-culture parameters. Subsequently, UACC-893, BT.20, or MDA.MB.453 were co-cultured with fibroblasts for 9 days. Co-cultures resulted in the generation of breast cancer histoid (BCH) with cancer cells showing the invasion of fibroblast spheroids, which were visualized by immunohistochemical (IHC) staining of sections (4 µm thick) of BCH. A reproducible quantitative expression of C-erbB.2 was detected in UACC-893 cancer cells in BCH sections by IHC staining and the Automated Cellular Imaging System. BCH sections also consistently exhibited qualitative expression of pancytokeratins, p53, Ki-67, or E-cadherin in cancer cells and that of vimentin or GSTPi in fibroblasts, fibronectin in the basement membrane and collagen IV in the extracellular matrix. The expression of the protein analytes and cellular architecture of BCH were markedly similar to those of breast cancer tissue.


Subject(s)
Breast Neoplasms/pathology , Bioreactors , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , Fibroblasts/cytology , Humans , Immunohistochemistry , Neoplasm Invasiveness , Receptor, ErbB-2/metabolism , Tumor Microenvironment
6.
Arch Pathol Lab Med ; 135(8): 1010-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21809992

ABSTRACT

CONTEXT: New guidelines for HER2 testing have been introduced. OBJECTIVES: To evaluate the difference in HER2 assessment after introduction of new cutoff levels for both immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) and to compare interobserver agreement and time to score between image analysis and conventional microscopy. DESIGN: Samples from 150 patients with breast cancer were scored by 7 pathologists using conventional microscopy, with a cutoff of both 10% and 30% IHC-stained cells, and using automated microscopy with image analysis. The IHC results were compared individually and to HER2 status as determined by FISH, using both the approved cutoff of 2.0 and the recently introduced cutoff of 2.2. RESULTS: High concordance was found in IHC scoring among the 7 pathologists. The 30% cutoff led to slightly fewer positive IHC observations. Introduction of a FISH equivocal zone affected 4% of the FISH scores. If cutoff for FISH is kept at 2.0, no difference in patient selection is found between the 10% and the 30% IHC cutoff. Among the 150 breast cancer samples, the new 30% IHC and 2.2 FISH cutoff levels resulted in one case without a firm diagnosis because both IHC and FISH were equivocal. Automated microscopy and image analysis-assisted IHC led to significantly better interobserver agreement among the 7 pathologists, with an increase in mean scoring time of only about 30 seconds per slide. CONCLUSIONS: The change in cutoff levels led to a higher concordance between IHC and FISH, but fewer samples were classified as HER2 positive.


Subject(s)
Image Processing, Computer-Assisted , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Receptor, ErbB-2 , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry/standards , In Situ Hybridization, Fluorescence/standards , Observer Variation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Reference Values , Reproducibility of Results
7.
J Immunol ; 185(9): 5607-18, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20876353

ABSTRACT

The T cell inhibitory ligand B7-H1 hinders T cell-mediated virus control, but also ameliorates clinical disease during autoimmune and virus-induced CNS disease. In mice infected with gliatropic demyelinating coronavirus, B7-H1 expression on oligodendroglia delays virus control, but also dampens clinical disease. To define the mechanisms by which B7-H1 alters pathogenic outcome, virus-infected B7-H1-deficient (B7-H1(-/-)) mice were analyzed for altered peripheral and CNS immune responses. B7-H1 deficiency did not affect peripheral T or B cell activation or alter the magnitude or composition of CNS-infiltrating cells. However, higher levels of IFN-γ mRNA in CNS-infiltrating virus-specific CD8 T cells as well as CD4 T cells contributed to elevated IFN-γ protein in the B7-H1(-/-) CNS. Increased effector function at the single-cell level was also evident by elevated granzyme B expression specifically in virus-specific CNS CD8 T cells. Although enhanced T cell activity accelerated virus control, 50% of mice succumbed to infection. Despite enhanced clinical recovery, surviving B7-H1(-/-) mice still harbored persisting viral mRNA, albeit at reduced levels compared with wild-type mice. B7-H1(-/-) mice exhibited extensive loss of axonal integrity, although demyelination, a hallmark of virus-induced tissue damage, was not increased. The results suggest that B7-H1 hinders viral control in B7-H1 expressing glia cells, but does not mediate resistance to CD8 T cell-mediated cytolysis. These data are the first, to our knowledge, to demonstrate that B7-H1-mediated protection from viral-induced immune pathology associated with encephalomyelitis resides in limiting T cell-mediated axonal bystander damage rather than direct elimination of infected myelinating cells.


Subject(s)
Axons/pathology , B7-1 Antigen/immunology , Bystander Effect/immunology , Encephalomyelitis/immunology , Membrane Glycoproteins/immunology , Peptides/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/immunology , B7-H1 Antigen , Cell Separation , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Cytokines/biosynthesis , Encephalomyelitis/pathology , Encephalomyelitis/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
8.
J Virol ; 84(10): 4878-88, 2010 May.
Article in English | MEDLINE | ID: mdl-20200240

ABSTRACT

Leukocyte access into the central nervous system (CNS) parenchyma is tightly regulated by the blood-brain barrier (BBB). Leukocyte migration through the endothelial cell wall into the perivascular space is well characterized; however, mechanisms regulating their penetration through the glia limitans into the parenchyma are less well studied, and the role of monocytes relative to neutrophils is poorly defined. Acute viral encephalitis was thus induced in CCL2-deficient (CCL2(-/-)) mice to specifically abrogate monocyte recruitment. Impaired monocyte recruitment prolonged T cell retention in the perivascular space, although no difference in overall CNS accumulation of CD4 or CD8 T cells was detected by flow cytometry. Delayed penetration to the CNS parenchyma was not associated with reduced or altered expression of either matrix metalloproteinases (MMP) or the T cell chemoattractants CXCL10 and CCL5. Nevertheless, decreased parenchymal leukocyte infiltration delayed T cell-mediated control of virus replication as well as clinical disease. These data are the first to demonstrate that the rapid monocyte recruitment into the CNS during viral encephalitis is dispensable for T cell migration across the blood vessel endothelium. However, monocytes facilitate penetration through the glia limitans. Thus, the rapid monocyte response to viral encephalitis constitutes an indirect antiviral pathway by aiding access of effector T cells to the site of viral infection.


Subject(s)
Blood-Brain Barrier/immunology , Encephalitis, Viral/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Animals , Cell Movement , Chemokine CCL2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/immunology
9.
J Virol ; 84(6): 3111-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042510

ABSTRACT

Neurotropic coronavirus induces acute encephalomyelitis and demyelination in mice. Infection of BALB/c (H-2(d)) mice expressing a dominant negative gamma interferon (IFN-gamma) receptor specifically in oligodendrocytes was examined to determine the influence of IFN-gamma signaling on pathogenesis. Inhibition of IFN-gamma signaling in oligodendrocytes increased viral load, infection of oligodendrocytes, oligodendrocyte loss, demyelination, and axonal damage resulting in increased mortality. IFN-gamma levels and the inflammatory response were not altered, although the level of tumor necrosis factor (TNF) mRNA was increased. These data indicate that IFN-gamma signaling by oligodendroglia reduces viral replication but affects both demyelination and tissue destruction in a host-specific manner.


Subject(s)
Coronavirus Infections/prevention & control , Encephalomyelitis/prevention & control , Interferon-gamma/immunology , Oligodendroglia/metabolism , Oligodendroglia/virology , Signal Transduction/immunology , Animals , Coronavirus Infections/immunology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Demyelinating Diseases/virology , Encephalomyelitis/immunology , Encephalomyelitis/pathology , Encephalomyelitis/virology , Gene Expression , Mice , Mice, Inbred BALB C , Murine hepatitis virus/immunology , Murine hepatitis virus/pathogenicity , Oligodendroglia/pathology , Receptors, Interferon/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Load , Virus Replication/immunology
10.
PLoS Pathog ; 5(10): e1000602, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19798426

ABSTRACT

IFN-alpha/beta plays a critical role in limiting viral spread, restricting viral tropism and protecting mice from neurotropic coronavirus infection. However, the IFN-alpha/beta dependent mechanisms underlying innate anti-viral functions within the CNS are poorly understood. The role of RNase L in viral encephalomyelitis was explored based on its functions in inhibiting translation, inducing apoptosis, and propagating the IFN-alpha/beta pathway through RNA degradation intermediates. Infection of RNase L deficient (RL(-/-)) mice with a sub-lethal, demyelinating mouse hepatitis virus variant revealed that the majority of mice succumbed to infection by day 12 p.i. However, RNase L deficiency did not affect overall control of infectious virus, or diminish IFN-alpha/beta expression in the CNS. Furthermore, increased morbidity and mortality could not be attributed to altered proinflammatory signals or composition of cells infiltrating the CNS. The unique phenotype of infected RL(-/-) mice was rather manifested in earlier onset and increased severity of demyelination and axonal damage in brain stem and spinal cord without evidence for enhanced neuronal infection. Increased tissue damage coincided with sustained brain stem infection, foci of microglia infection in grey matter, and increased apoptotic cells. These data demonstrate a novel protective role for RNase L in viral induced CNS encephalomyelitis, which is not reflected in overall viral control or propagation of IFN-alpha/beta mediated signals. Protective function is rather associated with cell type specific and regional restriction of viral replication in grey matter and ameliorated neurodegeneration and demyelination.


Subject(s)
Brain/pathology , Coronavirus Infections/enzymology , Demyelinating Diseases/pathology , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Endoribonucleases/metabolism , Animals , Apoptosis/physiology , Brain/enzymology , Brain/immunology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Encephalitis, Viral/pathology , Endoribonucleases/genetics , Endoribonucleases/immunology , Flow Cytometry , Fluorescent Antibody Technique , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Murine hepatitis virus , Nerve Degeneration/enzymology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/enzymology , Spinal Cord/immunology , Spinal Cord/pathology
11.
J Virol ; 83(12): 5978-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19339350

ABSTRACT

The relative contributions of interleukin-12 (IL-12) and IL-23 to viral pathogenesis have not been extensively studied. IL-12p40 mRNA rapidly increases after neurotropic coronavirus infection. Infection of mice defective in both IL-12 and IL-23 (p40(-/-)), in IL-12 alone (p35(-/-)), and in IL-23 alone (p19(-/-)) revealed that the symptoms of coronavirus-induced encephalitis are regulated by IL-12. IL-17-producing cells never exceeded background levels, supporting a redundant role of IL-23 in pathogenesis. Viral control, tropism, and demyelination were all similar in p35(-/-), p19(-/-), and wild-type mice. Reduced morbidity in infected IL-12 deficient mice was also not associated with altered recruitment or composition of inflammatory cells. However, gamma interferon (IFN-gamma) levels and virus-specific IFN-gamma-secreting CD4 and CD8 T cells were all reduced in the central nervous systems (CNS) of infected p35(-/-) mice. Transcription of the proinflammatory cytokines IL-1beta and IL-6, but not tumor necrosis factor, were initially reduced in infected p35(-/-) mice but increased to wild-type levels during peak inflammation. Furthermore, although transforming growth factor beta mRNA was not affected, IL-10 was increased in the CNS in the absence of IL-12. These data suggest that IL-12 does not contribute to antiviral function within the CNS but enhances morbidity associated with viral encephalitis by increasing the ratio of IFN-gamma to protective IL-10.


Subject(s)
Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Interleukin-12 Subunit p40/deficiency , Interleukin-23/deficiency , Animals , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/virology , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-12 Subunit p40/immunology , Interleukin-23/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Murine hepatitis virus/immunology
12.
J Immunol ; 182(9): 5430-8, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380790

ABSTRACT

The neurotropic coronavirus JHM strain of mouse hepatitis virus persists in oligodendroglia despite the presence of virus-specific CD8 T cells. Expression of programmed death 1 (PD-1) and B7-H1 were studied during acute and persistent infection to examine whether this negative regulatory mechanism contributes to CNS viral persistence. The majority of CNS-infiltrating CD8 T cells expressed PD-1, with the highest levels on virus-specific CD8 T cells. Moreover, despite control of infectious virus, CD8 T cells within the CNS of persistently infected mice maintained high PD-1 expression. Analysis of virus-susceptible target cells in vivo revealed that B7-H1 expression was regulated in a cell type-dependent manner. Oligodendroglia and microglia up-regulated B7-H1 following infection; however, although B7-H1 expression on oligodendroglia was prominent and sustained, it was significantly reduced and transient on microglia. Infection of mice deficient in the IFN-gamma or IFN-alpha/beta receptor demonstrated that B7-H1 expression on oligodendroglia is predominantly regulated by IFN-gamma. Ab blockade of B7-H1 on oligodendroglia in vitro enhanced IFN-gamma secretion by virus-specific CD8 T cells. More efficient virus control within the CNS of B7-H1-deficient mice confirmed inhibition of CD8 T cell function in vivo. Nevertheless, the absence of B7-H1 significantly increased morbidity without altering demyelination. These data are the first to demonstrate glia cell type-dependent B7-H1 regulation in vivo, resulting in adverse effects on antiviral CD8 T cell function. However, the beneficial role of PD-1:B7-H1 interactions in limiting morbidity highlights the need to evaluate tissue-specific intervention strategies.


Subject(s)
Antigens, Surface/physiology , Apoptosis Regulatory Proteins/physiology , B7-1 Antigen/physiology , Coronavirus Infections/mortality , Coronavirus Infections/prevention & control , Membrane Glycoproteins/physiology , Peptides/physiology , Spinal Cord Diseases/mortality , Spinal Cord Diseases/prevention & control , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , B7-1 Antigen/genetics , B7-H1 Antigen , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Line , Cell Movement/immunology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Murine hepatitis virus/immunology , Neuroglia/immunology , Neuroglia/pathology , Neuroglia/virology , Peptides/antagonists & inhibitors , Peptides/deficiency , Peptides/genetics , Programmed Cell Death 1 Receptor , Spinal Cord Diseases/immunology , Spinal Cord Diseases/pathology , Virus Replication/immunology
13.
Am J Pathol ; 173(4): 993-1001, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18772330

ABSTRACT

The endocannabinoid pathway plays an important role in the regulation of appetite and body weight, hepatic lipid metabolism, and fibrosis. Blockade of the endocannabinoid receptor CB1 with SR141716 promotes weight loss, reduces hepatocyte fatty acid synthesis, and is antifibrotic. D-4F, an apolipoprotein A-1 mimetic with antioxidant properties, is currently in clinical trials for the treatment of atherosclerosis. C57BL/6J mice were fed a high-fat diet for 7 months, followed by a 2.5-month treatment with either SR141716 or D-4F. SR141716 markedly improved body weight, liver weight, serum transaminases, insulin resistance, hyperglycemia, hypercholesterolemia, hyperleptinemia, and oxidative stress, accompanied by the significant prevention of fibrosis progression. D-4F improved hypercholesterolemia and hyperleptinemia without improvement in body weight, steatohepatitis, insulin resistance, or oxidative stress, and yet, there was significant prevention of fibrosis. D-4F prevented culture-induced activation of stellate cells in vitro. In summary, C57BL/6J mice given a high-fat diet developed features of metabolic syndrome with nonalcoholic steatohepatitis and fibrosis. Both SR141716 and D-4F prevented progression of fibrosis after onset of steatohepatitis, ie, a situation comparable to a common clinical scenario, with D-4F seeming to have a more general antifibrotic effect. Either compound therefore has the potential to be of clinical benefit.


Subject(s)
Fatty Liver/complications , Liver Cirrhosis/complications , Liver Cirrhosis/prevention & control , Metabolic Syndrome/complications , Actins/metabolism , Animals , Apolipoprotein A-I/pharmacology , Body Weight/drug effects , Cells, Cultured , Diet , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inflammation , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rimonabant
14.
J Virol ; 82(5): 2130-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094171

ABSTRACT

Replication of the neurotropic mouse hepatitis virus strain JHM (JHMV) is controlled primarily by CD8(+) T-cell effectors utilizing gamma interferon (IFN-gamma) and perforin-mediated cytotoxicity. CD4(+) T cells provide an auxiliary function(s) for CD8(+) T-cell survival; however, their direct contribution to control of virus replication and pathology is unclear. To examine a direct role of CD4(+) T cells in viral clearance and pathology, pathogenesis was compared in mice deficient in both perforin and IFN-gamma that were selectively reconstituted for these functions via transfer of virus-specific memory CD4(+) T cells. CD4(+) T cells from immunized wild-type, perforin-deficient, and IFN-gamma-deficient donors all initially reduced virus replication. However, prolonged viral control by IFN-gamma-competent donors suggested that IFN-gamma is important for sustained virus control. Local release of IFN-gamma was evident by up-regulation of class II molecules on microglia in recipients of IFN-gamma producing CD4(+) T cells. CD4(+) T-cell-mediated antiviral activity correlated with diminished clinical symptoms, pathology, and demyelination. Both wild-type donor CD90.1 and recipient CD90.2 CD4(+) T cells trafficked into the central nervous system (CNS) parenchyma and localized to infected white matter, correlating with decreased numbers of virus-infected oligodendrocytes in the CNS. These data support a direct, if limited, antiviral role for CD4(+) T cells early during acute JHMV encephalomyelitis. Although the antiviral effector mechanism is initially independent of IFN-gamma secretion, sustained control of CNS virus replication by CD4(+) T cells requires IFN-gamma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Central Nervous System Infections/immunology , Hepatitis, Viral, Animal/immunology , Murine hepatitis virus/immunology , Animals , Central Nervous System Infections/pathology , Central Nervous System Infections/virology , Flow Cytometry , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Murine hepatitis virus/physiology , Perforin/metabolism , Virus Replication
15.
J Virol ; 82(1): 300-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17928334

ABSTRACT

Neurotropic coronavirus infection induces expression of both beta interferon (IFN-beta) RNA and protein in the infected rodent central nervous system (CNS). However, the relative contributions of type I IFN (IFN-I) to direct, cell-type-specific virus control or CD8 T-cell-mediated effectors in the CNS are unclear. IFN-I receptor-deficient (IFNAR(-/-)) mice infected with a sublethal and demyelinating neurotropic virus variant and those infected with a nonpathogenic neurotropic virus variant both succumbed to infection within 9 days. Compared to wild-type (wt) mice, replication was prominently increased in all glial cell types and spread to neurons, demonstrating expanded cell tropism. Furthermore, increased pathogenesis was associated with significantly enhanced accumulation of neutrophils, tumor necrosis factor alpha, interleukin-6, chemokine (C-C motif) ligand 2, and IFN-gamma within the CNS. The absence of IFN-I signaling did not impair induction or recruitment of virus-specific CD8 T cells, the primary adaptive mediators of virus clearance in wt mice. Despite similar IFN-gamma-mediated major histocompatibility complex class II upregulation on microglia in infected IFNAR(-/-) mice, class I expression was reduced compared to that on microglia in wt mice, suggesting a synergistic role of IFN-I and IFN-gamma in optimizing class I antigen presentation. These data demonstrate a critical direct antiviral role of IFN-I in controlling virus dissemination within the CNS, even in the presence of potent cellular immune responses. By limiting early viral replication and tropism, IFN-I controls the balance of viral replication and immune control in favor of CD8 T-cell-mediated protective functions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System Diseases/immunology , Central Nervous System Diseases/virology , Coronavirus Infections/immunology , Interferon Type I/immunology , Animals , Central Nervous System/chemistry , Central Nervous System/virology , Central Nervous System Diseases/pathology , Chemokine CCL2/analysis , Histocompatibility Antigens Class I/biosynthesis , Interferon-gamma/analysis , Interleukin-6/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/virology , Neurons/virology , Neutrophils/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Survival Analysis , Tumor Necrosis Factor-alpha/analysis
16.
Neurosurg Focus ; 23(4): E13, 2007.
Article in English | MEDLINE | ID: mdl-17961037

ABSTRACT

Primary intraosseous meningiomas are a subtype of primary extradural meningiomas and constitute fewer than 2% of meningiomas overall, but they represent approximately two thirds of all extradural meningiomas. These types of meningiomas originate within the bones of the skull and thus can have a clinical presentation and radiographic differential diagnosis that is different from those for intradural meningiomas. Primary intraosseous meningiomas are classified based on their location and histopathological characteristics. Treatment primarily involves resection with wide margins if possible. Very little literature exists regarding the use of adjuvant therapies such as radiation and chemotherapy for these tumors. In fact, the literature regarding primary intra-osseous meningiomas consists mostly of clinical case reports and case series. This literature is reviewed and summarized in this article.


Subject(s)
Meningioma/diagnosis , Meningioma/therapy , Skull Neoplasms/diagnosis , Skull Neoplasms/therapy , Adult , Female , Humans , Meningioma/diagnostic imaging , Middle Aged , Radiography , Skull Neoplasms/diagnostic imaging
17.
Neurosurg Focus ; 23(4): E3, 2007.
Article in English | MEDLINE | ID: mdl-17961040

ABSTRACT

The histological appearance of a meningioma is an important predictor of tumor behavior and is frequently a factor in decisions concerning therapy. The relationship between histological features and prognosis is formalized in grading schemes such as those published by the World Health Organization (WHO), most recently in 2007. Although the latest edition is an improvement over previous grading schemes, the WHO scheme still fails to fully address a variety of important issues regarding the relationship between meningioma histological characteristics and behavior. In particular, routine histological examination fails to identify the subset of Grade I tumors that behave aggressively. Because of this, many additional prognostic markers that require immunohistochemical, cytogenetic, or molecular techniques to evaluate are under investigation. Only one, immunohistochemistry for the proliferation marker, Ki 67 (MIB-1), is used routinely and it has only limited utility. It is hoped that an understanding of the genetic changes that underlie tumor progression will improve healthcare professionals' ability to predict the behavior of meningiomas.


Subject(s)
Meningeal Neoplasms/pathology , Meningioma/pathology , Anaplasia/pathology , Antibodies, Antinuclear/metabolism , Antibodies, Monoclonal/metabolism , Humans , Meningeal Neoplasms/genetics , Meningeal Neoplasms/metabolism , Meningioma/genetics , Meningioma/metabolism , Neoplasm Invasiveness
18.
Int J Gynecol Pathol ; 26(3): 214-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17581401

ABSTRACT

The expression of the catalytic subunit of telomerase protein (human telomerase reverse transcriptase [hTERT]), which is associated with telomerase activity, was evaluated as a potential marker of the high-grade premalignant cervical intraepithelial neoplasia (CIN 2/3) lesions. For comparison, cases of normal cervical squamous mucosa, low-grade CIN1 lesion, and cervical squamous cell carcinoma were included. The hTERT expression was also compared with Ki-67 and topoisomerase II-alpha (TPII-alpha) to determine the proliferative activity of the hTERT-positive dysplastic cells by a quantitative immunohistochemical staining method and was classified as follows: negative, 5% or less; moderate, 6% to 50%; or high, greater than 50% of the positive cells. The hTERT-positive cells were detected in a patchy pattern in the lower parabasal layers and in much of the basal layer in normal squamous mucosa. A similar frequency of Ki-67- or TPII-alpha-positive cells was observed, with the exception of the basal layer cells that were mostly negative. It is worthy to note that the recognizable intact basal layer cells in cases of CIN lesions were also consistently positive for the expression of hTERT, but rarely for Ki-67 or TPII-alpha. The expression of hTERT was detected in a less patchy pattern at a high or moderate percentage of the dysplastic epithelial cells each in 28.5% of cases of CIN1 lesions. A similar frequency, high and moderate percentage combined, of the TPII-alpha-positive dysplastic cell was also observed. In contrast, a high percentage of the hTERT-positive dysplastic cells were detected as diffuse basal or full-length thickness in 87.5% or 95% of cases of CIN2 or CIN3, respectively. A similar frequency of Ki-67 or TPII-alpha expression was observed in the dysplastic cells of CIN3 lesions. The pattern of hTERT-positive malignant cells in squamous cell carcinoma and dysplastic cells in the high-grade CIN lesions, to a greater extent, and dysplastic cells in the low-grade CIN lesion, to a lesser extent, was distinct from that of the normal cervical squamous mucosa. The results suggest that the progressive increase in the hTERT expression, together with the proliferative activity of the dysplastic epithelial cells of the high-grade CIN lesions, represents an early genetic abnormality in cervical pathogenesis.


Subject(s)
Biomarkers, Tumor/biosynthesis , Telomerase/biosynthesis , Uterine Cervical Dysplasia/enzymology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology , Antigens, Neoplasm/biosynthesis , Cell Growth Processes/physiology , DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , Disease Progression , Female , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Retrospective Studies , Stromal Cells/enzymology , Stromal Cells/pathology , Telomerase/metabolism
19.
J Neurosci Res ; 84(6): 1225-34, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16955483

ABSTRACT

Agonists of liver X receptors (LXR), members of the nuclear hormone receptor superfamily, alter secretion of proinflammatory cytokines, suggesting potential antiinflammatory effects. A synthetic LXR agonist inhibited T-cell proliferation and cytokine release in a dose-dependent manner. Treatment of mice during induction of experimental autoimmune encephalomyelitis reduced clinical symptoms, central nervous system cellular inflammation, and major histocompatibility class II expression on microglia, as well as demyelination. In contrast to in vitro analysis, no reductions in peripheral neuroantigen specific T-cell responses were detected in comparing ligand and vehicle treated mice. These data suggest that LXR agonists play an important protective role in the regulation of T-cell-mediated inflammatory disease of the central nervous system.


Subject(s)
DNA-Binding Proteins/agonists , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Animals , CD4 Antigens/metabolism , Dose-Response Relationship, Drug , Female , Flow Cytometry , Gene Expression/drug effects , Genes, MHC Class II/genetics , Hydrocarbons, Fluorinated , Ligands , Liver X Receptors , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orphan Nuclear Receptors , Ovalbumin/immunology , Ovalbumin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
20.
Virology ; 350(1): 206-15, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16510164

ABSTRACT

Infection by the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in an acute encephalomyelitis associated with demyelination. T cells are critical in controlling viral replication, but also contribute to central nervous system (CNS) pathogenesis. To reveal a role for innate effectors in anti-viral immunity and neurological disease, JHMV pathogenesis was studied in mice deficient in interleukin-15 (IL-15-/-) and natural killer (NK) cells. Clinical disease, CNS inflammation and demyelination in infected IL-15-/- mice were similar to wild-type mice. Despite the absence of NK cells and suboptimal CD8+ T cell responses, IL-15-/- mice controlled JHMV replication as efficiently as wild-type mice. Similar kinetics of class I and class II upregulation on microglia further suggested no role of NK cells in regulating major histocompatibility complex (MHC) molecule expression on resident CNS cells. IL-15 and NK cells thus appear dispensable for anti-viral immunity and CNS pathogenesis during acute JHMV infection.


Subject(s)
Central Nervous System/pathology , Central Nervous System/virology , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Murine hepatitis virus/pathogenicity , Animals , CD8-Positive T-Lymphocytes/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/virology , Encephalomyelitis/immunology , Encephalomyelitis/virology , Gene Expression Regulation , Interleukin-15/deficiency , Interleukin-15/genetics , Major Histocompatibility Complex , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...