Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Am J Physiol Cell Physiol ; 322(3): C354-C369, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35044859

ABSTRACT

Suppressing mineralocorticoid receptor (MR) activity with MR antagonists is therapeutic for chronic skeletal muscle pathology in Duchenne muscular dystrophy (DMD) mouse models. Although mechanisms underlying clinical MR antagonist efficacy for DMD cardiomyopathy and other cardiac diseases are defined, mechanisms in skeletal muscles are not fully elucidated. Myofiber MR knockout improves skeletal muscle force and a subset of dystrophic pathology. However, MR signaling in myeloid cells is known to be a major contributor to cardiac efficacy. To define contributions of myeloid MR in skeletal muscle function and disease, we performed parallel assessments of muscle pathology, cytokine levels, and myeloid cell populations resulting from myeloid MR genetic knockout in muscular dystrophy and acute muscle injury. Myeloid MR knockout led to lower levels of C-C motif chemokine receptor 2 (CCR2)-expressing macrophages, resulting in sustained myofiber damage after acute injury of normal muscle. In acute injury, myeloid MR knockout also led to increased local muscle levels of the enzyme that produces the endogenous MR agonist aldosterone, further supporting important contributions of MR signaling in normal muscle repair. In muscular dystrophy, myeloid MR knockout altered cytokine levels differentially between quadriceps and diaphragm muscles, which contain different myeloid populations. Myeloid MR knockout led to higher levels of fibrosis in dystrophic diaphragm. These results support important contributions of myeloid MR signaling to skeletal muscle repair in acute and chronic injuries and highlight the useful information gained from cell-specific genetic knockouts to delineate mechanisms of pharmacological efficacy.


Subject(s)
Diaphragm/metabolism , Macrophages/metabolism , Muscular Diseases/metabolism , Muscular Dystrophy, Duchenne/metabolism , Quadriceps Muscle/metabolism , Receptors, Mineralocorticoid/metabolism , Aldosterone/metabolism , Animals , Barium Compounds , Chlorides , Cytokines/genetics , Cytokines/metabolism , Diaphragm/immunology , Diaphragm/pathology , Disease Models, Animal , Female , Fibrosis , Macrophages/immunology , Male , Mice, Inbred mdx , Mice, Knockout , Muscular Diseases/chemically induced , Muscular Diseases/immunology , Muscular Diseases/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Quadriceps Muscle/immunology , Quadriceps Muscle/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Mineralocorticoid/genetics , Signal Transduction
3.
PLoS One ; 13(3): e0194421, 2018.
Article in English | MEDLINE | ID: mdl-29561896

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle weakness which is ultimately fatal, most often due to involvement of the diaphragm. Macrophage infiltration of dystrophic muscles has been strongly linked to muscle damage and fibrosis in DMD. We hypothesized that cenicriviroc (CVC), a dual chemokine receptor (CCR2/CCR5) antagonist currently under clinical evaluation for other diseases, could prevent macrophage accumulation and blunt disease progression in the diaphragms of mdx mice (genetic homologue of DMD). Treatment with CVC (20 mg/kg/day intraperitoneally) or vehicle was initiated in mdx mice at 2 weeks of age (prior to the onset of muscle necrosis) and continued for 4 weeks. Flow cytometry to assess inflammatory cell subsets as well as histological and force generation parameters were determined in mdx diaphragms at the conclusion of the treatment. CVC therapy induced a major (3.9-fold) reduction in total infiltrating macrophages, whereas total numbers of neutrophils and T lymphocytes (CD4+ and CD8+) were unaffected. No changes in macrophage polarization status (inflammatory versus anti-inflammatory skewing based on iNOS and CD206 expression) were observed. Muscle fiber size and fibrosis were not altered by CVC, whereas a significant reduction in centrally nucleated fibers was found suggesting a decrease in prior necrosis-regeneration cycles. In addition, maximal isometric force production by the diaphragm was increased by CVC therapy. These results suggest that CVC or other chemokine receptor antagonists which reduce pathological macrophage infiltration may have the potential to slow disease progression in DMD.


Subject(s)
Imidazoles/pharmacology , Macrophages/immunology , Muscular Dystrophy, Duchenne/drug therapy , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR5 , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Diaphragm/immunology , Diaphragm/pathology , Isometric Contraction/drug effects , Isometric Contraction/immunology , Macrophages/pathology , Mice , Mice, Inbred mdx , Muscle Strength/drug effects , Muscle Strength/immunology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Receptors, CCR2/immunology , Severity of Illness Index , Sulfoxides
4.
J Pathol ; 244(3): 323-333, 2018 03.
Article in English | MEDLINE | ID: mdl-29214629

ABSTRACT

Chronic muscle inflammation is a critical feature of Duchenne muscular dystrophy and contributes to muscle fibre injury and disease progression. Although previous studies have implicated T cells in the development of muscle fibrosis, little is known about their role during the early stages of muscular dystrophy. Here, we show that T cells are among the first cells to infiltrate mdx mouse dystrophic muscle, prior to the onset of necrosis, suggesting an important role in early disease pathogenesis. Based on our comprehensive analysis of the kinetics of the immune response, we further identify the early pre-necrotic stage of muscular dystrophy as the relevant time frame for T-cell-based interventions. We focused on protein kinase C θ (PKCθ, encoded by Prkcq), a critical regulator of effector T-cell activation, as a potential target to inhibit T-cell activity in dystrophic muscle. Lack of PKCθ not only reduced the frequency and number of infiltrating T cells but also led to quantitative and qualitative changes in the innate immune cell infiltrate in mdx/Prkcq-/- muscle. These changes were due to the inhibition of T cells, since PKCθ was necessary for T-cell but not for myeloid cell infiltration of acutely injured muscle. Targeting T cells with a PKCθ inhibitor early in the disease process markedly diminished the size of the inflammatory cell infiltrate and resulted in reduced muscle damage. Moreover, diaphragm necrosis and fibrosis were also reduced following treatment. Overall, our findings identify the early T-cell infiltrate as a therapeutic target and highlight the potential of PKCθ inhibition as a therapeutic approach to muscular dystrophy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Diaphragm/drug effects , Muscular Dystrophy, Animal/prevention & control , Protein Kinase C-theta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/drug effects , Animals , Diaphragm/enzymology , Diaphragm/immunology , Diaphragm/pathology , Disease Models, Animal , Fibrosis , Immunity, Innate/drug effects , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Necrosis , Protein Kinase C-theta/deficiency , Protein Kinase C-theta/genetics , Protein Kinase C-theta/metabolism , Severity of Illness Index , Signal Transduction/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Time Factors
5.
Biomaterials ; 77: 320-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26618750

ABSTRACT

The currently available surgical options to repair the diaphragm are associated with significant risks of defect recurrence, lack of growth potential and restored functionality. A tissue engineered diaphragm has the potential to improve surgical outcomes for patients with congenital or acquired disorders. Here we show that decellularized diaphragmatic tissue reseeded with bone marrow mesenchymal stromal cells (BM-MSCs) facilitates in situ regeneration of functional tissue. A novel bioreactor, using simultaneous perfusion and agitation, was used to rapidly decellularize rat diaphragms. The scaffolds retained architecture and mechanical properties and supported cell adhesion, proliferation and differentiation. Biocompatibility was further confirmed in vitro and in vivo. We replaced 80% of the left hemidiaphragm with reseeded diaphragmatic scaffolds. After three weeks, transplanted animals gained 32% weight, showed myography, spirometry parameters, and histological evaluations similar to native rats. In conclusion, our study suggested that reseeded decellularized diaphragmatic tissue appears to be a promising option for patients in need of diaphragmatic reconstruction.


Subject(s)
Diaphragm/transplantation , Mesenchymal Stem Cell Transplantation/methods , Tissue Engineering/methods , Tissue Scaffolds , Absorbable Implants , Allografts , Animals , Bioreactors , Cell Adhesion , Cell Differentiation , Diaphragm/blood supply , Diaphragm/diagnostic imaging , Diaphragm/immunology , Electromyography , Graft Survival , Hernias, Diaphragmatic, Congenital , Macrophages/immunology , Male , Neovascularization, Physiologic , Radiography , Rats , Rats, Inbred Lew , Tissue Engineering/instrumentation , Transplantation, Heterotopic , Transplants/blood supply , Transplants/immunology , Transplants/physiology , Wound Healing
6.
Prog Brain Res ; 212: 221-35, 2014.
Article in English | MEDLINE | ID: mdl-25194200

ABSTRACT

Acute intermittent hypoxia (AIH) triggers a form of respiratory plasticity known as long-term facilitation (LTF), which is manifested as a progressive increase in respiratory motor activity that lasts for minutes to hours after the hypoxic stimulus is removed. Respiratory LTF has been reported in numerous animal models, but it appears to be influenced by a variety of factors (e.g., species, age, and gender). While most studies focusing on respiratory LTF have been conducted in adult (including young adult) rat preparations, little is known about the influence of postnatal maturation on AIH-induced respiratory LTF. To begin to address this issue, we examined diaphragm EMG activity in response to and at 5-min intervals for 60 min following three 5-min episodes of hypoxia (8% O2) in urethane-anesthetized spontaneously breathing P14-P15 neonatal rats (n=15). For these experiments, the hypoxic episodes were separated by hyperoxia (40% O2), and all rats were continuously supplied with ~4% CO2. During the AIH trials, burst frequency was increased by ~20-90% above baseline in each of the rats examined while changes in burst amplitude were highly variable. Following the AIH episodes, respiratory LTF was characterized by predominantly an increase in burst frequency (fLTF) ranging from ~10% to 55%, with most rats exhibiting a 20-40% increase. In seven rats, however, an increase in amplitude (ampLTF) (~10%, n=3; ~20%, n=3; ~30%, n=1) was also noted. These data suggest that in contrast to observations in anesthetized ventilated adult rats, in anesthetized spontaneously breathing P14-P15 neonatal rats, respiratory LTF is dominated by fLTF, not ampLTF.


Subject(s)
Hypoxia/physiopathology , Long-Term Potentiation/physiology , Phrenic Nerve/physiology , Respiratory Mechanics/physiology , Anesthetics, Intravenous , Animals , Animals, Newborn , Diaphragm/immunology , Diaphragm/physiology , Electromyography , Rats , Rats, Sprague-Dawley , Urethane
7.
PLoS One ; 8(9): e73457, 2013.
Article in English | MEDLINE | ID: mdl-24039949

ABSTRACT

Diaphragmatic contractility is reduced in preterm lambs after lipopolysaccharide (LPS) exposure in utero. The mechanism of impaired fetal diaphragm contractility after LPS exposure is unknown. We hypothesise that in utero exposure to LPS induces a deficiency of mitochondrial complex activity and oxidative damage in the fetal diaphragm. To test this hypothesis, we used a well-established preterm ovine model of chorioamnionitis: Pregnant ewes received intra-amniotic (IA) saline or 10 mg LPS, at 2 d or 7 d prior to surgical delivery at 121 d GA (term = 150 d). The fetus was killed humanely immediately after delivery for tissue sampling. Mitochondrial fractions were prepared from the isolated diaphragm and mitochondrial electron transfer chain activities were evaluated using enzymatic assays. Oxidative stress was investigated by quantifying mitochondrial oxidative protein levels and determining antioxidant gene and protein (catalase, superoxide dismutase 2 and glutathione peroxidase 1) expression. The activity of the erythroid 2-related factor 2 (Nrf2)-mediated antioxidant signalling pathway was examined by quantifying the Nrf2 protein content of cell lysate and nuclear extract. A 2 d LPS exposure in utero significantly decreased electron transfer chain complex II and IV activity (p<0.05). A 7 d LPS exposure inhibited superoxide dismutase 2 and catalase expression at gene and protein levels, and Nrf2 pathway activity (p<0.05) compared with control and 2 d LPS groups, respectively. Diaphragm mitochondria accumulated oxidised protein after a 7 d LPS exposure. We conclude that intrauterine exposure to LPS induces mitochondrial oxidative stress and electron chain dysfunction in the fetal diaphragm, that is further exacerbated by impairment of the antioxidant signalling pathway and decreased antioxidant activity.


Subject(s)
Chorioamnionitis/immunology , Diaphragm/embryology , Diaphragm/immunology , Fetus/immunology , Lipopolysaccharides/immunology , Oxidative Stress , Animals , Diaphragm/physiology , Electron Transport , Female , Mitochondria/immunology , Pregnancy , Sheep
8.
Sci Rep ; 3: 2183, 2013.
Article in English | MEDLINE | ID: mdl-23851606

ABSTRACT

The molecular mechanism of muscle degeneration in a lethal muscle disorder Duchene muscular dystrophy (DMD) has not been fully elucidated. The dystrophic dog, a model of DMD, shows a high mortality rate with a marked increase in serum creatine kinase (CK) levels in the neonatal period. By measuring serum CK levels in cord and venous blood, we found initial pulmonary respiration resulted in massive diaphragm damage in the neonates and thereby lead to the high serum CK levels. Furthermore, molecular biological techniques revealed that osteopontin was prominently upregulated in the dystrophic diaphragm prior to the respiration, and that immediate-early genes (c-fos and egr-1) and inflammation/immune response genes (IL-6, IL-8, COX-2, and selectin E) were distinctly overexpressed after the damage by the respiration. Hence, we segregated dystrophic phases at the molecular level before and after mechanical damage. These molecules could be biomarkers of muscle damage and potential targets in pharmaceutical therapies.


Subject(s)
Creatine Kinase/blood , Diaphragm/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Respiration , Animals , Animals, Newborn , Diaphragm/immunology , Diaphragm/metabolism , Disease Models, Animal , Dogs , Gene Expression Regulation , Hyalin/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Molecular Sequence Annotation , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Osteopontin/genetics , Osteopontin/metabolism , Proteolysis , Respiration/genetics , Signal Transduction , Transcription, Genetic , Transcriptome
9.
Am J Respir Cell Mol Biol ; 49(5): 866-74, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23795611

ABSTRACT

Preterm birth is associated with inflammation of the fetal membranes (chorioamnionitis). We aimed to establish how chorioamnionitis affects the contractile function and phenotype of the preterm diaphragm. Pregnant ewes received intra-amniotic injections of saline or 10 mg LPS, 2 days or 7 days before delivery at 121 days of gestation (term = 150 d). Diaphragm strips were dissected for the assessment of contractile function after terminal anesthesia. The inflammatory cytokine response, myosin heavy chain (MHC) fibers, proteolytic pathways, and intracellular molecular signaling were analyzed using quantitative PCR, ELISA, immunofluorescence staining, biochemical assays, and Western blotting. Diaphragm peak twitch force and maximal tetanic force were approximately 30% lower than control values in the 2-day and 7-day LPS groups. Activation of the NF-κB pathway, an inflammatory response, and increased proteasome activity were observed in the 2-day LPS group relative to the control or 7-day LPS group. No inflammatory response was evident after a 7-day LPS exposure. Seven-day LPS exposure markedly decreased p70S6K phosphorylation, but no effect on other signaling pathways was evident. The proportion of MHC IIa fibers was lower than that for control samples in the 7-day LPS group. MHC I fiber proportions did not differ between groups. These results demonstrate that intrauterine LPS impairs preterm diaphragmatic contractility after 2-day and 7-day exposures. Diaphragm dysfunction, resulting from 2-day LPS exposure, was associated with a transient activation of proinflammatory signaling, with subsequent increased atrophic gene expression and enhanced proteasome activity. Persistently impaired contractility for the 7-day LPS exposure was associated with the down-regulation of a key component of the protein synthetic signaling pathway and a reduction in the proportions of MHC IIa fibers.


Subject(s)
Chorioamnionitis/physiopathology , Diaphragm/physiopathology , Lipopolysaccharides , Myocardial Contraction , Animals , Chorioamnionitis/blood , Chorioamnionitis/chemically induced , Chorioamnionitis/immunology , Cytokines/metabolism , Diaphragm/immunology , Diaphragm/metabolism , Disease Models, Animal , Female , Gestational Age , Inflammation Mediators/blood , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/metabolism , Muscle Strength , Muscular Atrophy/blood , Muscular Atrophy/immunology , Muscular Atrophy/physiopathology , Myosin Heavy Chains/blood , NF-kappa B/metabolism , Pregnancy , Proteasome Endopeptidase Complex/metabolism , Sheep , Signal Transduction , Time Factors
10.
Muscle Nerve ; 46(5): 705-10, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23055311

ABSTRACT

INTRODUCTION: We investigated the localization of a ganglioside, N-acetylgalactosaminyl GD1a (GalNAc-GD1a), in peripheral nerves with an IgG anti-GalNAc-GD1a antibody, which was produced in rabbits immunized with GalNAc-GD1a. METHODS: Teased fibers from ventral and dorsal roots and hemidiaphragm sections of rats were assessed using fluorescent double- and triple-labeling methods. RESULTS: The nodal and paranodal regions of teased fibers from ventral roots were immunostained with IgG anti-GalNAc-GD1a antibodies. After collagenase treatment, no staining was seen with IgG anti-GalNAc-GD1a or anti-NF200 antibodies, whereas α-bungarotoxin selectively stained nerve terminals. In cross-sectional and longitudinal sections of rat hemidiaphragm, IgG anti-GalNAc-GD1a antibodies overlapped with α-BuTx and anti-NF200 antibodies, indicating that GalNAc-GD1a is localized to the nerve terminal. IgG anti-GalNAc-GD1a antibody staining also overlapped with that of AChR clusters and syntaxin-positive presynaptic nerve terminals. CONCLUSION: GalNAc-GD1 is localized in both pre- and postsynaptic nerve terminals of neuromuscular junctions.


Subject(s)
Binding Sites, Antibody , Diaphragm/metabolism , Gangliosides/immunology , Gangliosides/metabolism , Immunoglobulin G/metabolism , Neuromuscular Junction/metabolism , Animals , Diaphragm/chemistry , Diaphragm/immunology , Female , Neuromuscular Junction/chemistry , Neuromuscular Junction/immunology , Protein Binding/immunology , Rabbits , Rats , Rats, Wistar
11.
J Appl Physiol (1985) ; 113(10): 1594-603, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22961265

ABSTRACT

Resistive breathing (encountered in chronic obstructive pulmonary disease and asthma) results in cytokine upregulation and decreased nitric oxide (NO) levels in the strenuously contracting diaphragm. NO can regulate gene expression. We hypothesized that endogenously produced NO downregulates cytokine production triggered by strenuous diaphragmatic contraction. Wistar rats treated with vehicle, the nonselective NO synthase inhibitor NG-nitro-l-arginine-methylester (l-NAME), or the NO donor diethylenetriamine-NONOate (DETA) were subjected to inspiratory resistive breathing (IRB; 50% of maximal inspiratory pressure) for 6 h or sham operation. Additional groups of rats were subjected to IRB for 6 h with concurrent administration of l-NAME and inhibitors of NF-κB (BAY-11-7082), ERK1/2 (PD98059), or P38 (SB203580). Inhibition of NO production (with l-NAME) resulted in upregulation of IRB-induced diaphragmatic IL-6, IL-10, IL-2, TNF-α, and IL-1ß levels by 50%, 53%, 60%, 47%, and 45%, respectively. In contrast, the NO donor (DETA) attenuated the IRB-induced cytokine upregulation to levels characteristic of quietly breathing animals. l-NAME augmented IRB-induced activation of MAPKs (P38 and ERK1/2) and NF-κB, whereas DETA triggered the opposite effect. NF-κB and ERK1/2 inhibition in l-NAME-treated animals blunted the l-NAME-induced cytokine upregulation except IL-6, whereas P38 inhibition blunted all (including IL-6) cytokine upregulation. NO downregulates IRB-induced cytokine production in the strenuously contracting diaphragm through its action on MAPKs and NF-κB.


Subject(s)
Cytokines/metabolism , Diaphragm/metabolism , Inflammation/metabolism , Inhalation , Lung Diseases, Obstructive/metabolism , Muscle Contraction , Nitric Oxide/metabolism , Animals , Diaphragm/drug effects , Diaphragm/immunology , Enzyme Inhibitors/pharmacology , Inflammation/immunology , Inflammation/physiopathology , Lung Diseases, Obstructive/immunology , Lung Diseases, Obstructive/physiopathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Protein Carbonylation , Rats , Rats, Wistar , Signal Transduction , Time Factors , Work of Breathing
12.
Foodborne Pathog Dis ; 9(1): 75-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22011040

ABSTRACT

Serum and meat juice analyses for the detection of Toxoplasma gondii antibodies by an immunofluorescence antibody assay were compared in 100 seropositive and 100 seronegative slaughtered heavy swine. Meat juice was obtained from diaphragm and gracilis muscles of the serologically tested animals. Seventy-two diaphragmatic meat juice samples (36%, 95% interval confidence [IC] 29.4%-43.1%) and 63 gracilis meat juice samples (31.5%, 95% IC 25.1%-38.4%) tested positive for T. gondii antibodies. The average concordance between serum and meat juice derived from both muscles was "substantial" (K=0.6-0.8). The K-value was 1 when considering serum samples showing a titer >1/16, whereas it decreased to 0.62 and to 0.49 when considering serum samples with a 1/16 titer in meat juice from diaphragm and gracilis muscle, respectively.


Subject(s)
Antibodies, Protozoan/blood , Fluorescent Antibody Technique/methods , Meat/analysis , Swine Diseases/diagnosis , Toxoplasma/immunology , Toxoplasmosis, Animal/diagnosis , Abattoirs , Animals , Chlorocebus aethiops , Diaphragm/immunology , Diaphragm/parasitology , Enzyme-Linked Immunosorbent Assay , Meat/parasitology , Muscle, Skeletal/immunology , Muscle, Skeletal/parasitology , Reference Standards , Sensitivity and Specificity , Swine , Swine Diseases/blood , Swine Diseases/parasitology , Toxoplasma/isolation & purification , Toxoplasmosis, Animal/blood , Toxoplasmosis, Animal/parasitology , Vero Cells
13.
Am J Physiol Regul Integr Comp Physiol ; 300(5): R1152-62, 2011 May.
Article in English | MEDLINE | ID: mdl-21325641

ABSTRACT

Inspiratory resistive breathing (IRB) induces cytokine expression in the diaphragm. The mechanism of this cytokine induction remains elusive. The roles of MAPKs and NF-κB and the impact of oxidative stress in IRB-induced cytokine upregulation in the diaphragm were studied. Wistar rats were subjected to IRB (50% of maximal inspiratory pressure) via a two-way nonrebreathing valve for 1, 3, or 6 h. Additional groups of rats subjected to IRB for 6 h were randomly assigned to receive either solvent or N-acetyl-cysteine (NAC) or inhibitors of NF-κB (BAY-11-7082), ERK1/2 (PD98059), and P38 MAPK (SB203580) to study the effect of oxidative stress, NF-κB, and MAPKs in IRB-induced cytokine upregulation in the diaphragm. Quietly breathing animals served as controls. IRB upregulated cytokine (IL-6, TNF-α, IL-10, IL-2, IL-1ß) protein levels in the diaphragm and resulted in increased activation of MAPKs (P38, ERK1/2) and NF-κB. Inhibition of NF-κB and ERK1/2 blunted the upregulation of all cytokines except that of IL-6, which was further increased. P38 inhibition attenuated all cytokine (including IL-6) upregulation. Both P38 and ERK1/2 inhibition decreased NF-κB/p65 subunit phosphorylation. NAC pretreatment blunted IRB-induced cytokine upregulation in the diaphragm and resulted in decreased ERK1/2, P38, and NF-κB/p65 phosphorylation. In conclusion, IRB-induced cytokine upregulation in the diaphragm is under the regulatory control of MAPKs and NF-κB. IL-6 is regulated differently from all other cytokines through a P38-dependent and NF-κB independent pathway. Oxidative stress is a stimulus for IRB-induced cytokine upregulation in the diaphragm.


Subject(s)
Airway Resistance , Cytokines/metabolism , Diaphragm/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Inhalation , NF-kappa B/metabolism , Oxidative Stress , Work of Breathing , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Blood Gas Analysis , Diaphragm/drug effects , Diaphragm/immunology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Oxidative Stress/drug effects , Phosphorylation , Pressure , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Time Factors , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
PLoS Genet ; 5(7): e1000586, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19649303

ABSTRACT

Cystic fibrosis (CF) patients often have reduced mass and strength of skeletal muscles, including the diaphragm, the primary muscle of respiration. Here we show that lack of the CF transmembrane conductance regulator (CFTR) plays an intrinsic role in skeletal muscle atrophy and dysfunction. In normal murine and human skeletal muscle, CFTR is expressed and co-localized with sarcoplasmic reticulum-associated proteins. CFTR-deficient myotubes exhibit augmented levels of intracellular calcium after KCl-induced depolarization, and exposure to an inflammatory milieu induces excessive NF-kB translocation and cytokine/chemokine gene upregulation. To determine the effects of an inflammatory environment in vivo, sustained pulmonary infection with Pseudomonas aeruginosa was produced, and under these conditions diaphragmatic force-generating capacity is selectively reduced in Cftr(-/-) mice. This is associated with exaggerated pro-inflammatory cytokine expression as well as upregulation of the E3 ubiquitin ligases (MuRF1 and atrogin-1) involved in muscle atrophy. We conclude that an intrinsic alteration of function is linked to the absence of CFTR from skeletal muscle, leading to dysregulated calcium homeostasis, augmented inflammatory/atrophic gene expression signatures, and increased diaphragmatic weakness during pulmonary infection. These findings reveal a previously unrecognized role for CFTR in skeletal muscle function that may have major implications for the pathogenesis of cachexia and respiratory muscle pump failure in CF patients.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Animals , Calcium/metabolism , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cytokines/immunology , Diaphragm/immunology , Diaphragm/metabolism , Diaphragm/pathology , Diaphragm/physiopathology , Disease Models, Animal , Gene Expression , Genetic Predisposition to Disease , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muscle Weakness , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology
15.
Hum Mol Genet ; 17(15): 2280-92, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18430716

ABSTRACT

The immune response to dystrophin-deficient muscle promotes the pathology of Duchenne muscular dystrophy (DMD) and the mdx mouse model of DMD. In this investigation, we find that the release of major basic protein (MBP) by eosinophils is a prominent feature of DMD and mdx dystrophy and that eosinophils lyse muscle cells in vitro by the release of MBP-1. We also show that eosinophil depletions of mdx mice by injections of anti-chemokine receptor-3 reduce muscle cell lysis, although lysis of mdx muscle membranes is not reduced by null mutation of MBP-1 in vivo. However, ablation of MBP-1 expression in mdx mice produces other effects on muscular dystrophy. First, fibrosis of muscle and hearts, a major cause of mortality in DMD, is greatly reduced by null mutation of MBP-1 in mdx mice. Furthermore, either ablation of MBP-1 or eosinophil depletion causes large increases in cytotoxic T-lymphocytes (CTLs) in mdx muscles. The increase in CTLs in MBP-1-null mice does not reflect a general shift toward a Th1 inflammatory response, because the mutation had no significant effect on the expression of interferon-gamma, inducible nitric oxide synthase or tumor necrosis factor. Rather, MBP-1 reduces the activation and proliferation of splenocytes in vitro, indicating that MBP-1 acts in a more specific immunomodulatory role to affect the inflammatory response in muscular dystrophy. Together, these findings show that eosinophil-derived MBP-1 plays a significant role in regulating muscular dystrophy by attenuating the cellular immune response and promoting tissue fibrosis that can eventually contribute to increased mortality.


Subject(s)
Eosinophil Major Basic Protein/physiology , Eosinophils/immunology , Muscles/pathology , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Animals , Antibodies, Monoclonal/pharmacology , Diaphragm/immunology , Diaphragm/pathology , Disease Models, Animal , Eosinophil Major Basic Protein/genetics , Fibrosis , Humans , Leukocyte Reduction Procedures , Mice , Mice, Inbred mdx , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscles/immunology , Muscular Dystrophy, Duchenne/genetics , Mutation , Myocardium/immunology , Myocardium/pathology , Receptors, CCR3/antagonists & inhibitors , Regeneration/genetics , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology
16.
Neurobiol Dis ; 28(1): 113-21, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17720506

ABSTRACT

Acute motor axonal neuropathy (AMAN) variant of Guillain-Barré syndrome is often associated with IgG anti-GM1 and -GD1a antibodies. The pathophysiological basis of antibody-mediated selective motor nerve dysfunction remains unclear. We investigated the effects of IgG anti-GM1 and -GD1a monoclonal antibodies (mAbs) on neuromuscular transmission and calcium influx in hemidiaphragm preparations and in cultured neurons, respectively, to elucidate mechanisms of Ab-mediated muscle weakness. Anti-GM1 and -GD1a mAbs depressed evoked quantal release to a significant yet different extent, without affecting postsynaptic currents. At equivalent concentrations, anti-GD1b, -GT1b, or sham mAbs did not affect neuromuscular transmission. At fourfold higher concentration, an anti-GD1b mAb (specificity described in immune sensory neuropathies) induced completely reversible blockade. In neuronal cultures, anti-GM1 and -GD1a mAbs significantly reduced depolarization-induced calcium influx. In conclusion, different anti-ganglioside mAbs induce distinct effects on presynaptic transmitter release by reducing calcium influx, suggesting that this is one mechanism of antibody-mediated muscle weakness in AMAN.


Subject(s)
Autoantibodies/metabolism , Gangliosides/immunology , Neurons/metabolism , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , Animals , Antibodies, Monoclonal , Autoantibodies/immunology , Autoantigens/immunology , Calcium , Cells, Cultured , Diaphragm/immunology , Diaphragm/metabolism , Mice , Mice, Inbred BALB C , Neuromuscular Junction/immunology , Neuromuscular Junction/physiology , Neurons/immunology , Neurotransmitter Agents/metabolism , Olfactory Bulb , Organ Culture Techniques , Patch-Clamp Techniques , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/physiopathology , Presynaptic Terminals/immunology
17.
Am J Respir Cell Mol Biol ; 36(4): 504-12, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17122368

ABSTRACT

Severe weakness of the respiratory muscles, with attendant respiratory failure and death, has been documented in sepsis. In this study, we show that during murine pulmonary infection with Pseudomonas aeruginosa, multiple proinflammatory genes are up-regulated not only within the lungs, but also within the diaphragm. Significant induction of TNF-alpha, IL-1alpha, IL-1beta, IL-6, and IL-18 gene expression occurred within the diaphragm in a bacterial dose-dependent manner. We determined whether the anti-inflammatory cytokine IL-10 could blunt proinflammatory gene expression within the diaphragm under these conditions. The IL-10 receptor was found to be expressed by the diaphragm in vivo as well as in primary diaphragmatic muscle cell cultures. Transduction of myoblasts with an adenoviral vector (Ad-IL-10) induced strong IL-10 expression, and intramuscular injection of the same vector in vivo produced significant increases in IL-10 serum levels. Ad-IL-10 treatment of mice infected with P. aeruginosa significantly inhibited the induction of proinflammatory cytokines within the diaphragm, but not in the infected lungs. Ad-IL-10 treatment also led to greatly improved diaphragmatic force production in infected mice. These results suggest that pulmonary infection triggers proinflammatory gene expression by the diaphragm along with diaphragmatic weakness. Shifting the balance between pro- and anti-inflammatory mediators in favor of the latter by IL-10 gene delivery was able to restore normal diaphragmatic force-generating capacity under these conditions, suggesting a possible avenue for therapeutic intervention.


Subject(s)
Cytokines/metabolism , Diaphragm/immunology , Interleukin-10/physiology , Lung Diseases/immunology , Muscle Contraction , Pseudomonas Infections/immunology , Animals , Cells, Cultured , Diaphragm/microbiology , Diaphragm/physiopathology , Inflammation/immunology , Inflammation/microbiology , Lung Diseases/microbiology , Mice , Mice, Inbred C57BL , Receptors, Interleukin-10/metabolism , Transduction, Genetic , Up-Regulation
18.
Neurobiol Dis ; 23(3): 697-707, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16837207

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease leading to motor neuron cell death, but recent studies suggest that non-neuronal cells may contribute to the pathological mechanisms involved. Myostatin is a negative regulator of muscle growth whose function can be inhibited using neutralizing antibodies. In this study, we used transgenic mouse and rat models of ALS to test whether treatment with anti-myostatin antibody slows muscle atrophy, motor neuron loss, or disease onset and progression. Significant increases in muscle mass and strength were observed in myostatin-antibody-treated SOD1(G93A) mice and rats prior to disease onset and during early-stage disease. By late stage disease, only diaphragm muscle remained significantly different in treated animals in comparison to untreated controls. Myostatin inhibition did not delay disease onset nor extend survival in either the SOD1(G93A) mouse or rat. Together, these results indicate that inhibition of myostatin does not protect against the onset and progression of motor neuron degenerative disease. However, the preservation of skeletal muscle during early-stage disease and improved diaphragm morphology and function maintained through late stage disease suggest that anti-myostatin therapy may promote some improved muscle function in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Antibodies/pharmacology , Growth Inhibitors/antagonists & inhibitors , Muscle, Skeletal/physiopathology , Muscular Atrophy/therapy , Transforming Growth Factor beta/antagonists & inhibitors , Age of Onset , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Animals, Genetically Modified , Antibodies/immunology , Antibodies/therapeutic use , Cell Death/drug effects , Cell Death/physiology , Diaphragm/immunology , Diaphragm/innervation , Diaphragm/physiopathology , Disease Models, Animal , Female , Growth Inhibitors/immunology , Growth Inhibitors/metabolism , Humans , Male , Mice , Mice, Knockout , Motor Neurons/immunology , Motor Neurons/pathology , Muscle Weakness/immunology , Muscle Weakness/physiopathology , Muscle Weakness/therapy , Muscle, Skeletal/immunology , Muscle, Skeletal/innervation , Muscular Atrophy/immunology , Muscular Atrophy/physiopathology , Myostatin , Organ Size/drug effects , Organ Size/immunology , Rats , Recovery of Function/drug effects , Recovery of Function/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Survival Rate , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Treatment Outcome
19.
J Neuroimmunol ; 175(1-2): 77-86, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16647144

ABSTRACT

Irreversible connective tissue proliferation in muscle is a pathological hallmark of Duchenne muscular dystrophy (DMD), a genetic degenerative muscle disease due to lack of the sarcolemmal protein dystrophin. Focal release of transforming growth factor-beta1 (TGF-beta1) is involved in fibrosis development. Murine muscular dystrophy (mdx) is genetically homologous to DMD and histopathological alterations comparable to those in DMD muscles occur in diaphragm of older mdx mice. To investigate the early development of fibrosis and TGF-beta1 involvement, we assessed diaphragms in 6-36-week-old mdx and C57/BL6 (control) mice for fibrosis, and used real-time PCR and ELISA to determine TGF-beta1 expression. Significantly greater fibrosis and TGF-beta1 expression were found in mdx from the 6th week. Mice treated with neutralizing antibody against TGF-beta1 had lower levels of TGF-beta1 protein, reduced fibrosis, unchanged muscles fiber degeneration/regeneration, but increased inflammatory cells (CD4+lymphocytes). These data demonstrate early and progressive fibrosis in mdx diaphragm accompanied by TGF-beta1 upregulation. Reduction of TGF-beta1 appears promising as a therapeutic approach to muscle fibrosis, but further studies are required to evaluate long term effects of TGF-beta1 immunomodulation on the immune system.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cell Proliferation , Connective Tissue/immunology , Connective Tissue/pathology , Diaphragm/immunology , Diaphragm/pathology , Immunologic Factors/therapeutic use , Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Fibrosis/immunology , Fibrosis/pathology , Fibrosis/prevention & control , Growth Inhibitors/biosynthesis , Growth Inhibitors/immunology , Immunoglobulin G/administration & dosage , Immunoglobulin G/therapeutic use , Immunologic Factors/administration & dosage , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta1
20.
Mov Disord ; 20(12): 1617-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16078216

ABSTRACT

With the advent of a commercial preparation of botulinum toxin type B (BT-B) for treatment of cervical dystonia detection of antibodies against BT-B (BT-B-AB) becomes necessary. For this purpose, we carried out a mouse diaphragm assay (MDA) by continuous measurement of the twitch force of a mouse hemidiaphragm preparation elicited by electric stimulation of its phrenic nerve. After exposing the preparation to BT-B 3 ng/ml the time to half-maximal twitch force reduction (paralysis time [PT]) was 69 +/- 4 min (n = 25). Addition of sera from patients with antibodies against BT-A produced a PT of 68 +/- 5 min (n = 24), whereas addition of sera from controls with antibodies against tetanus toxoid produced a PT of 67 +/- 6 min (n = 30). When defined amounts of BT-B-AB were added to the MDA, PT was prolonged. This prolongation was correlated closely to the amount of BT-B-AB added, thus producing a calibration curve. The threshold for BT-B-AB detection was 0.4 mU/ml. When sera from 7 patients (4 women, 3 men; age 50.6 +/- 14.2 years) with cervical dystonia (Toronto Western Spasmodic Torticollis Rating Scale score, 18.9 +/- 2.9) and complete secondary failure of BT-B therapy (NeuroBloc; Elan Pharmaceuticals, Shannon, Ireland; 12,229 +/- 2,601 MU/injection series, 1.86 +/- 0.69 injection series before complete secondary therapy failure; 100.4 +/- 15.8 days between injection series with normal therapeutic effect) were tested, BT-B-AB titers of more than 10 mU/ml were found in all of them. The MDA can be used to measure neutralizing BT-B-AB titers quantitatively and with adequate sensitivity and specificity. Further studies are necessary to understand the role of intermediate BT-B-AB titers in partial BT-B therapy failure.


Subject(s)
Anti-Dyskinesia Agents/immunology , Antibodies/analysis , Botulinum Toxins/immunology , Diaphragm/immunology , Torticollis/immunology , Animals , Anti-Dyskinesia Agents/therapeutic use , Biological Assay/methods , Botulinum Toxins/therapeutic use , Botulinum Toxins, Type A , Diaphragm/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Male , Mice , Middle Aged , Time Factors , Torticollis/blood , Torticollis/drug therapy , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...