Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Dis Esophagus ; 25(2): 166-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21819482

ABSTRACT

The pathogenesis of eosinophilic esophagitis (EoE) is incompletely understood. In certain eosinophilic diseases, activation of tyrosine kinase after fusion of the Fip1-like-1 and platelet-derived growth factor receptor-α genes (F-P fusion gene) mediates eosinophilia via downstream effectors such as extracellular-regulated kinase (ERK1/2) and signal transducers and activators of transcription (STAT5). This mechanism has not been examined in EoE. Our aim was to detect the F-P fusion gene, pERK1/2, and pSTAT5 in esophageal tissue from patients with EoE, gastroesophageal reflux disease (GERD), and normal controls. We performed a cross-sectional pilot study comparing patients with steroid-responsive and steroid-refractory EoE, to GERD patients and normal controls. EoE cases were defined by consensus guidelines. Fluorescence in situ hybridization (FISH) was performed to detect the F-P fusion gene and immunohistochemistry (IHC) was performed to detect pERK1/2 and pSTAT5 in esophageal biopsies. Twenty-nine subjects (median age 30 years [range 1-59]; 16 males; 24 Caucasians) were included: eight normal, six GERD, and 15 EoE (five steroid-refractory). On FISH, 98%, 99%, and 99% of the nuclei in the normal, GERD, and EoE groups, respectively, were normal (P= 0.42). On IHC, a median of 250, 277, and 479 nuclei/mm(2) stained for pERK 1/2 in the normal, GERD, and EoE groups, respectively (P= 0.07); the refractory EoE patients had the highest degree pERK 1/2 staining (846 nuclei/mm(2); P= 0.07). No trend was seen for pSTAT5. In conclusion, the F-P fusion gene was not detected with increased frequency in EoE. Patients with EoE had a trend toward higher levels of pERK 1/2, but not STAT5, in the esophageal epithelium, with highest levels in steroid-refractory EoE patients.


Subject(s)
Eosinophilic Esophagitis/metabolism , Gastroesophageal Reflux/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , STAT5 Transcription Factor/metabolism , Adolescent , Adult , Biomarkers/metabolism , Child , Child, Preschool , Cross-Sectional Studies , Eosinophilic Esophagitis/genetics , Female , Gastroesophageal Reflux/genetics , Humans , In Situ Hybridization, Fluorescence , Infant , MAP Kinase Signaling System/physiology , Male , Middle Aged , Pilot Projects , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Retrospective Studies , STAT5 Transcription Factor/genetics , Young Adult
2.
Oncogene ; 29(34): 4787-99, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20562910

ABSTRACT

Topoisomerase IIalpha (topoIIalpha) is an essential mammalian enzyme that topologically modifies DNA and is required for chromosome segregation during mitosis. Previous research suggests that inhibition of topoII decatenatory activity triggers a G(2) checkpoint response, which delays mitotic entry because of insufficient decatenation of daughter chromatids. Here we examine the effects of both topoIIalpha and topoIIbeta on decatenatory activity in cell extracts, DNA damage and decatenation G(2) checkpoint function, and the frequencies of p16(INK4A) allele loss and gain. In diploid human fibroblast lines, depletion of topoIIalpha by small-interfering RNA was associated with severely reduced decatenatory activity, delayed progression from G(2) into mitosis and insensitivity to G(2) arrest induced by the topoII catalytic inhibitor ICRF-193. Furthermore, interphase nuclei of topoIIalpha-depleted cells showed increased frequencies of losses and gains of the tumor suppressor genetic locus p16(INK4A). This study shows that the topoIIalpha protein is required for decatenation G(2) checkpoint function, and inactivation of decatenation and the decatenation G(2) checkpoint leads to abnormal chromosome segregation and genomic instability.


Subject(s)
Antigens, Neoplasm/physiology , DNA Topoisomerases, Type II/physiology , DNA-Binding Proteins/physiology , G2 Phase/genetics , Genomic Instability , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromosome Segregation/physiology , DNA/biosynthesis , DNA/genetics , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diketopiperazines , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , G2 Phase/drug effects , Genes, p16 , Humans , Mitosis/drug effects , Mitosis/genetics , Piperazines/pharmacology , Signal Transduction/genetics , Topoisomerase II Inhibitors
3.
Infect Immun ; 69(12): 7729-35, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705954

ABSTRACT

We investigated the importance of the host complement system in the pathogenesis of disease mediated by the intramacrophage pathogen Mycobacterium avium. Mycobacteria opsonized with complement are efficiently ingested by macrophages through various complement receptors. Furthermore, unlike other bacteria, mycobacteria can activate both the alternative and classical complement pathways in the absence of specific antibodies. Therefore, to examine the role of complement in the mycobacterial infection process in vivo, mice deficient in complement component C3 were infected with M. avium. Surprisingly, C3-deficient mice infected intravenously with M. avium displayed no difference in bacterial burden or granulomatous response compared to wild-type control mice. C3-sufficient mice and C3-deficient mice were equally susceptible to infection by M. avium regardless of the genotype at the bcg locus, a locus known to confer susceptibility to infection with intracellular pathogens. In vitro studies using mouse bone marrow-derived macrophages resulted in significant M. avium invasion of macrophages in the absence of C3; however, the kinetics of infection were delayed compared to complement-mediated invasion. The data indicate that complement does not play an essential role in mediating M. avium infections in the mouse and suggest either that other invasion mechanisms can compensate for the absence of complement-mediated entry or that complement is not a major mycobacterial opsonin in vivo.


Subject(s)
Complement C3/deficiency , Mycobacterium avium/immunology , Tuberculosis/etiology , Tuberculosis/immunology , Animals , Antibodies, Bacterial , Antibody Specificity , Bone Marrow Cells/immunology , Complement Pathway, Alternative , Complement Pathway, Classical , Disease Susceptibility , Granuloma/etiology , Liver/microbiology , Liver/pathology , Macrophages/immunology , Mice , Mice, Mutant Strains , Opsonin Proteins , Spleen/microbiology , Tumor Necrosis Factor-alpha/biosynthesis
4.
Muscle Nerve ; 24(5): 695-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11317281

ABSTRACT

Cell-based therapies, such as myoblast transfer therapy, are likely to become an integral part of any approach to treat myopathies such as Duchenne muscular dystrophy. Previous studies have shown that an increased level of regeneration in the host muscle enhances incorporation of donor myoblasts. Leukemia inhibitory factor (LIF) increases the number of dystrophic fibers expressing dystrophin after myoblast transplantation and enhances regeneration in injured and diseased muscle. Morphometric analysis was used to investigate whether an increased level of regeneration is induced by LIF after myoblast transplantation. We found that, in muscles treated with LIF, the number of fibers undergoing regeneration was increased. The increased incorporation of donor myoblasts and thus dystrophin expression induced by LIF may be due, at least in part, to an increased level of regeneration of dystrophic muscle.


Subject(s)
Growth Inhibitors/pharmacology , Interleukin-6 , Lymphokines/pharmacology , Muscle, Skeletal/physiology , Regeneration/drug effects , Animals , Cell Transplantation , Dystrophin/biosynthesis , Leukemia Inhibitory Factor , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/transplantation , Muscle, Skeletal/cytology , Transplantation, Heterologous
5.
Muscle Nerve ; 23(11): 1700-5, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11054748

ABSTRACT

Although the muscles of the mdx mouse lack dystrophin, the protein absent in muscles of humans affected with Duchenne muscular dystrophy (DMD), the only mdx muscle to degenerate in a manner similar to those of DMD boys is the diaphragm. We have previously shown that leukemia inhibitory factor (LIF) is a trauma factor that enhances muscle repair in vivo and, when applied exogenously, increases the fiber size of mdx skeletal muscle. Furthermore, we developed a controlled release device for LIF based on a calcium alginate rod (release rate about 0.5% per day). These rods were sutured to the abdominal surface of the hemidiaphragm of mdx mice 3 months old. At age 6 months the mice were killed and the diaphragm muscles fixed and sectioned. The sections showed obvious muscle degeneration at 3 months of age in mdx mouse diaphragms and further degeneration at 6 months in saline-perfused muscle. Hemidiaphragm muscles continuously exposed to LIF over the same period contained more normal myofibers, larger regenerated fibers, and less adipose tissue and other non-contractile tissue. Morphometric analysis of the diaphragm sections was carried out. The LIF-treated animals showed a significant increase in fiber number and size compared to saline rod controls. The amount of nonmuscle (connective tissue and adipose tissue) was significantly reduced and the maximum force-producing capacity of isolated diaphragm muscle strips was higher in LIF-treated mice. The results demonstrate that LIF treatment ameliorates the dystrophic abnormalities in mdx mouse diaphragm.


Subject(s)
Growth Inhibitors/pharmacology , Interleukin-6 , Lymphokines/pharmacology , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/pathology , Animals , Cell Size/drug effects , Diaphragm/cytology , Diaphragm/pathology , Diaphragm/physiology , Infusion Pumps, Implantable , Leukemia Inhibitory Factor , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Fatigue/drug effects , Recombinant Proteins/pharmacology
6.
J Neurol Sci ; 160 Suppl 1: S106-13, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9851659

ABSTRACT

Growth factors are theoretically promising agents for ALS therapy, but have been disappointing in subcutaneous delivery due to either toxicity or lack of major efficacy. Leukaemia inhibitory factor (LIF), was named after its effect on haemopoietic cells, and belongs to a group of cytokines which includes CNTF, IL-6, CT-1, OM and IL-11. All group members use the gp130 signal transducing subunit for intracellular signalling, but show differences in biological effect. In vitro and in vivo studies on axotomy and nerve crush models demonstrate a powerful effect of LIF in the survival of both motor and sensory neurones, while reducing denervation induced muscle atrophy. Its effects in muscle also include stimulating myoblast proliferation in vitro, and up-regulation after muscle injury. LIF will also stimulate muscle regeneration in vivo when applied exogenously after injury. In published studies of both axotomy induced neuronal death and in the Wobbler mouse models LIF is active at doses of 10 microg/kg delivered systemically, well below the expected maximum tolerated dose suggested by primate safety studies. LIF is expressed in low levels by spinal cord neurones with significant up-regulation when the neurones are damaged by BOAA toxin, an excitatory amino acid associated with a form of ALS. This augments other evidence suggesting LIF is a trauma factor playing a role in the injury response of adult neuronal tissue, and may be more effective than related growth factors. Taken together, the data suggests LIF is a physiologically relevant trophic factor with implications in clinical medicine as a therapy for ALS, and a human recombinant form (AM424), entered human clinical trials during 1998.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Growth Inhibitors/therapeutic use , Interleukin-6 , Lymphokines/therapeutic use , Animals , Disease Models, Animal , Drug Delivery Systems , Growth Inhibitors/chemistry , Growth Inhibitors/physiology , Humans , Leukemia Inhibitory Factor , Lymphokines/chemistry , Lymphokines/physiology , Mice , Mice, Neurologic Mutants , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Nervous System/drug effects , Neurons/drug effects , Receptors, Cytokine/metabolism , Regeneration/drug effects , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase-1
7.
Neurosci Lett ; 249(1): 1-4, 1998 Jun 12.
Article in English | MEDLINE | ID: mdl-9672374

ABSTRACT

A model of spinal trauma was developed where spinal neurones of adult mice were exposed to the excitotoxic glutamate analogue beta-N-oxylamino-L-alanine (L-BOAA). After 24 h, the injured neurones received a single dose of [125I]-LIF at the same site of the spinal cord, and 2 h later, tissues were removed to assess the distribution of leukaemia inhibitory factor (LIF). There was a significant increase in LIF binding to the injured region of the spinal cord over saline controls, and this corresponded with a significant increase in LIF mRNA expression in the same region of the cord. There was a change in the expression of ciliary neurotrophic factor, but the expression of cardiotrophin-1 (CT-1) and the common receptor subunit LIF receptor beta (LIFRbeta) did not change after neurotoxin treatment. The results add to the evidence that LIF plays a significant role in the response of adult neuronal tissue to injury.


Subject(s)
Amino Acids, Diamino , Growth Inhibitors/biosynthesis , Interleukin-6 , Lymphokines/biosynthesis , Motor Neuron Disease/metabolism , Spinal Diseases/metabolism , Animals , Ciliary Neurotrophic Factor , Cytokines/biosynthesis , Growth Inhibitors/metabolism , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Motor Neuron Disease/chemically induced , Motor Neuron Disease/pathology , Motor Neurons/metabolism , Nerve Tissue Proteins/biosynthesis , Neurotoxins/toxicity , RNA, Messenger/biosynthesis , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/metabolism , Receptors, OSM-LIF , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Diseases/chemically induced , Spinal Diseases/pathology , Up-Regulation , beta-Alanine/analogs & derivatives , beta-Alanine/toxicity
8.
Muscle Nerve ; 20(7): 815-22, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9179153

ABSTRACT

Although a number of cytokines have been implicated in tissue regeneration, it is unknown which ones actually function in vivo. Here, we use mice with a targeted mutation in the leukemia inhibitory factor (LIF) gene to examine the role of LIF in muscle regeneration. Using a muscle crush model, we show that muscle regeneration in LIF knockout mice is significantly, reduced compared to control littermates. Further, targeted infusion of LIF in both normal and LIF knockout animals stimulated muscle regeneration, but the stimulation observed was much greater in the mutant animals than in controls. In contrast, interleukin-6 and transforming growth factor-alpha, which also stimulate myoblast proliferation in vitro, had no effect on regeneration. These findings demonstrate directly that LIF is involved in regeneration of injured muscle and points to the use of LIF as a therapeutic agent in the treatment of neuromuscular disease.


Subject(s)
Growth Inhibitors/pharmacokinetics , Lymphokines/pharmacokinetics , Muscle, Skeletal/physiology , Regeneration/drug effects , Animals , Cell Division/drug effects , Cell Size/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Growth Inhibitors/genetics , Interleukin-6/pharmacokinetics , Iodine Radioisotopes , Leukemia Inhibitory Factor , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Recombinant Proteins/pharmacology , Regeneration/genetics , Transforming Growth Factor alpha/pharmacokinetics
10.
Growth Factors ; 15(1): 61-8, 1997.
Article in English | MEDLINE | ID: mdl-9401818

ABSTRACT

Leukaemia inhibitory factor (LIF) has been shown to effectively enhance skeletal muscle regeneration after mechanical injury and it may have potential therapeutic use in the muscular dystrophies as well as peripheral nerve repair after injury. When LIF is applied systemically to an animal, it is rapidly removed with a biological half life of only a few minutes, and at high doses it exhibits toxic effects. Calcium alginate rods have been developed for the purpose of insertion adjacent to skeletal muscles. These rods, when charged with LIF will release the growth factor to the muscle at a rate of less than 1% per day and for a period extending to several months. In addition, tubes of alginate are described which will be suitable for the continuous supply of LIF to repaired peripheral nerve.


Subject(s)
Alginates , Drug Carriers , Growth Inhibitors/pharmacokinetics , Interleukin-6 , Lymphokines/pharmacokinetics , Muscle, Skeletal/metabolism , Animals , Dystrophin/metabolism , Glucuronic Acid , Growth Inhibitors/pharmacology , Growth Inhibitors/therapeutic use , Hexuronic Acids , Iodine Radioisotopes , Leukemia Inhibitory Factor , Lymphokines/pharmacology , Lymphokines/therapeutic use , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Polylysine/pharmacology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...