Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Mol Ther Methods Clin Dev ; 15: 305-319, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31890730

ABSTRACT

rAAVrh74.MCK.GALGT2 is a surrogate gene therapy that inhibits muscular dystrophy in multiple animal models. Here, we report on a dose-response study of functional muscle GALGT2 expression as well as toxicity and biodistribution studies after systemic intravenous (i.v.) delivery of rAAVrh74.MCK.GALGT2. A dose of 4.3 × 1014vg/kg (measured with linear DNA standard) resulted in GALGT2-induced glycosylation in the majority of skeletal myofibers throughout the body and in almost all cardiomyocytes, while several lower doses also showed significant muscle glycosylation. No adverse clinical signs or treatment-dependent changes in tissue or organ pathology were noted at 1 or 3 months post-treatment. Blood cell and serum enzyme chemistry measures in treated mice were all within the normal range except for alkaline phosphatase (ALP) activity, which was elevated in serum but not in tissues. Some anti-rAAVrh74 capsid T cell responses were noted at 4 weeks post-treatment, but all such responses were not present at 12 weeks. Using intramuscular delivery, GALGT2-induced muscle glycosylation was increased in Cmah-deficient mice, which have a humanized sialoglycome, relative to wild-type mice, suggesting that use of mice may underestimate GALGT2 activity in human muscle. These data demonstrate safety and high transduction of muscles throughout the body plan with i.v. delivery of rAAVrh74.MCK.GALGT2.

2.
Mol Ther Methods Clin Dev ; 10: 89-104, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30073180

ABSTRACT

Recombinant adeno-associated virus (rAAV)rh74.MCK.GALGT2 is a muscle-specific gene therapy that is being developed to treat forms of muscular dystrophy. Here we report on an isolated limb infusion technique in a non-human primate model, where hindlimb blood flow is transiently isolated using balloon catheters to concentrate vector in targeted leg muscles. A bilateral dose of 2.5 × 1013 vector genomes (vg)/kg/limb was sufficient to induce GALGT2-induced glycosylation in 10%-60% of skeletal myofibers in all leg muscles examined. There was a 19-fold ± 6-fold average limb-wide increase in vector genomes per microgram genomic DNA at a bilateral dose of 2.5 × 1013 vg/kg/limb compared with a bilateral dose of 6 × 1012 vg/kg/limb. A unilateral dose of 6 × 1013 vg/kg/limb showed a 12- ± 3-fold increase in treated limb muscles compared to contralateral untreated limb muscles, which received vector only after release into the systemic circulation from the treated limb. Variability in AAV biodistribution between different segments of the same muscle was 125% ± 18% for any given dose, while variability between the same muscle for any given treatment dose was 45% ± 7%. These experiments demonstrate that treatment of muscles throughout the leg with rAAVrh74.MCK.GALGT2 can be accomplished safely using an isolated limb infusion technique, where balloon catheters transiently isolate the limb vasculature, but that intra- and inter-muscle transduction variability is a significant issue.

3.
Hum Gene Ther ; 28(6): 493-509, 2017 06.
Article in English | MEDLINE | ID: mdl-28345428

ABSTRACT

Use of adeno-associated virus (AAV) to transduce genes into skeletal muscles can be associated with T-cell responses to viral capsid and/or to transgenic protein. Intramuscular mononuclear cell infiltrates primarily consisting of CD8+ T cells and also containing FOXP3+ regulatory T cells were present in rhesus macaque skeletal muscle treated with rAAVrh74.MCK.GALGT2 by vascular delivery. Administration of oral prednisone prior to AAV gene delivery and throughout the study reduced such infiltrates by 60% at 24 weeks post AAV delivery compared with AAV-treated animals not receiving prednisone, regardless of the presence of pre-existing AAV serum antibodies at the time of treatment. The majority of CD8+ T cells in AAV-treated muscles expressed activated caspase 3 and programmed cell death protein 1 (PD1), suggesting ongoing programmed cell death. AAV-transduced skeletal muscles also had elevated expression of programmed death ligand 2 (PDL2) on skeletal myofibers, and this increase in expression extended to muscles where transgene was not overexpressed. These data demonstrate that prednisone can reduce the extent of intramuscular T-cell infiltrates in AAV-treated muscles, which may aid in achieving long-term transgene expression, as may the induction of PDL2 expression on skeletal myofibers to promote PD1-mediated programmed T-cell death.


Subject(s)
Dependovirus/genetics , Genetic Vectors/immunology , Immunosuppressive Agents/pharmacology , Muscular Dystrophies/therapy , Prednisone/pharmacology , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Receptor/genetics , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Death , Dependovirus/immunology , Gene Expression , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Immunity, Cellular/drug effects , Injections, Intra-Arterial , Injections, Intramuscular , Macaca mulatta , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/immunology , Muscular Dystrophies/pathology , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , Programmed Cell Death 1 Ligand 2 Protein/agonists , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/agonists , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transgenes
4.
J Neuromuscul Dis ; 3(2): 247-260, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27854211

ABSTRACT

BACKGROUND: Duchenne Muscular Dystrophy (DMD) is a severe, progressive, neuromuscular disorder of childhood. While a number of serum factors have been identified as potential biomarkers of DMD, none, as yet, are proteins within the dystrophin-associated glycoprotein (DAG) complex. OBJECTIVE: We have developed an immobilized serum ELISA assay to measure the expression of a constitutively cleaved and secreted component of the DAG complex, the N-terminal domain of α dystroglycan (αDG-N), and assayed relative expression in serum from muscular dystrophy patients and normal controls. METHODS: ELISAs of immobilized patient or mouse serum and Western blots were used to assess αDG-N expression. RESULTS: Immobilization of diluted serum on ELISA plates was important for this assay, as methods to measure serum αDG-N in solution were less robust. αDG-N ELISA signals were significantly reduced in DMD serum (27±3% decrease, n = 9, p < 0.001) relative to serum from otherwise normal controls (n = 38), and calculated serum αDG-N concentrations were reduced in DMD relative to normal (p < 0.01) and Becker Muscular Dystrophy (n = 11, p < 0.05) patient serum. By contrast, ELISA signals from patients with Inclusion Body Myositis were not different than normal (4±3% decrease, n = 8, p = 0.99). αDG-N serum signals were also significantly reduced in utrophin-deficient mdx mice as compared to mdx and wild type mice. CONCLUSIONS: Our results are the first demonstration of a component of the DAG complex as a potential serum biomarker in DMD. Such a serum measure could be further developed as a tool to help reflect overall muscle DAG complex expression or stability.


Subject(s)
Dystroglycans/blood , Muscular Dystrophy, Duchenne/blood , Animals , Biomarkers/blood , Blotting, Western , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Animal/blood , Myositis, Inclusion Body/blood , Utrophin/genetics
5.
Mol Ther ; 22(4): 713-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24145553

ABSTRACT

Overexpression of GALGT2 in skeletal muscle can stimulate the glycosylation of α dystroglycan and the upregulation of normally synaptic dystroglycan-binding proteins, some of which are dystrophin and laminin α2 surrogates known to be therapeutic for several forms of muscular dystrophy. This article describes the vascular delivery of GALGT2 gene therapy in a large animal model, the rhesus macaque. Recombinant adeno-associated virus, rhesus serotype 74 (rAAVrh74), was used to deliver GALGT2 via the femoral artery to the gastrocnemius muscle using an isolated focal limb perfusion method. GALGT2 expression averaged 44 ± 4% of myofibers after treatment in macaques with low preexisting anti-rAAVrh74 serum antibodies, and expression was reduced to 9 ± 4% of myofibers in macaques with high preexisting rAAVrh74 immunity (P < 0.001; n = 12 per group). This was the case regardless of the addition of immunosuppressants, including prednisolone, tacrolimus, and mycophenolate mofetil. GALGT2-treated macaque muscles showed increased glycosylation of α dystroglycan and increased expression of dystrophin and laminin α2 surrogate proteins, including utrophin, plectin1, agrin, and laminin α5. These experiments demonstrate successful transduction of rhesus macaque muscle with rAAVrh74.MCK.GALGT2 after vascular delivery and induction of molecular changes thought to be therapeutic in several forms of muscular dystrophy.


Subject(s)
Dystrophin/biosynthesis , Gene Transfer Techniques , Genetic Therapy , Laminin/biosynthesis , Muscular Dystrophies/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dystroglycans/genetics , Dystroglycans/metabolism , Dystrophin/genetics , Gene Expression Regulation , Glycosyltransferases/genetics , Laminin/genetics , Macaca mulatta/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Muscular Dystrophies/therapy
6.
J Cardiovasc Magn Reson ; 15: 94, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107555

ABSTRACT

BACKGROUND: Patients with non-ST-segment elevation acute coronary syndrome (NSTE-ACS) have varying degrees of salvageable myocardium at risk of irreversible injury. We hypothesized that a novel model of NSTE-ACS produces acute myocardial injury, measured by increased T2 cardiovascular magnetic resonance (CMR), without significant necrosis by late gadolinium enhancement (LGE). METHODS: In a canine model, partial coronary stenosis was created and electrodes placed on the epicardium. Myocardial T2, an indicator of at-risk myocardium, was measured pre- and post-tachycardic pacing. RESULTS: Serum troponin-I (TnI) was not detectable in unoperated sham animals but averaged 1.97 ± 0.72 ng/mL in model animals. Coronary stenosis and pacing produced significantly higher T2 in the affected vs. the remote myocardium (53.2 ± 4.9 vs. 43.6 ± 2.8 ms, p < 0.01) with no evident injury by LGE. Microscopy revealed no significant irreversible cellular injury. Relative respiration rate (RRR) of affected vs. remote myocardial tissue was significantly lower in model vs. sham animals (0.72 ± 0.07 vs. 1.04 ± 0.07, p < 0.001). Lower RRR corresponded to higher final TnI levels (R(2) = 0.83, p = 0.004) and changes in CaMKIID and mitochondrial gene expression. CONCLUSIONS: A large animal NSTE-ACS model with mild TnI elevation and without ST elevation, similar to the human syndrome, demonstrates signs of acute myocardial injury by T2-CMR without significant irreversible damage. Reduced tissue respiration and associated adaptations of critical metabolic pathways correspond to increased myocardial injury by serum biomarkers in this model. T2-CMR as a biomarker of at-risk but salvageable myocardium warrants further consideration in preclinical and clinical studies of NSTE-ACS.


Subject(s)
Acute Coronary Syndrome/diagnosis , Magnetic Resonance Imaging , Myocardium/pathology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/genetics , Acute Coronary Syndrome/pathology , Acute Coronary Syndrome/physiopathology , Animals , Biomarkers/blood , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Respiration , Disease Models, Animal , Dogs , Gene Expression Regulation , Genes, Mitochondrial , Myocardium/metabolism , Necrosis , Organs at Risk , Oxygen Consumption , Predictive Value of Tests , Reproducibility of Results , Stroke Volume , Time Factors , Tissue Survival , Troponin I/blood , Ventricular Function, Left
7.
PLoS One ; 8(8): e72354, 2013.
Article in English | MEDLINE | ID: mdl-23951313

ABSTRACT

OBJECTIVE: Mitochondrial transcription factor A (TFAM) is normally bound to and remains associated with mitochondrial DNA (mtDNA) when released from damaged cells. We hypothesized that TFAM, bound to mtDNA (or equivalent CpG-enriched DNA), amplifies TNFα release from TLR9-expressing plasmacytoid dendritic cells (pDCs) by engaging RAGE. MATERIALS AND METHODS: Murine Flt3 ligand-expanded splenocytes obtained from C57BL/6 mice were treated with recombinant human TFAM, alone or in combination with CpG-enriched DNA with subsequent TNFα release measured by ELISA. The role of RAGE was determined by pre-treatment with soluble RAGE or heparin or by employing matching RAGE (-/-) splenocytes. TLR9 signaling was evaluated using a specific TLR9-blocking oligonucleotide and by inhibiting endosomal processing, PI3K and NF-κB. Additional studies examined whether heparin sulfate moieties or endothelin converting enzyme-1 (ECE-1)-dependent recycling of endosomal receptors were required for TFAM and CpG DNA recognition. MAIN RESULTS: TFAM augmented splenocyte TNFα release in response to CpGA DNA, which was strongly dependent upon pDCs and regulated by RAGE and TLR9 receptors. Putative TLR9 signaling pathways, including endosomal acidification and signaling through PI3K and NF-κB, were essential for splenocyte TNFα release in response to TFAM+CpGA DNA. Interestingly, TNFα release depended upon endothelin converting enzyme (ECE)-1, which cleaves and presumably activates TLR9 within endosomes. Recognition of the TFAM-CpGA DNA complex was dependent upon heparin sulfate moieties, and recombinant TFAM Box 1 and Box 2 proteins were equivalent in terms of augmenting TNFα release. CONCLUSIONS: TFAM promoted TNFα release in a splenocyte culture model representing complex cell-cell interactions in vivo with pDCs playing a critical role. To our knowledge, this study is the first to incriminate ECE-1-dependent endosomal cleavage of TLR9 as a critical step in the signaling pathway leading to TNFα release. These findings, and others reported herein, significantly advance our understanding of sterile immune responses triggered by mitochondrial danger signals.


Subject(s)
DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Toll-Like Receptor 9/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , CpG Islands , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Dendritic Cells/immunology , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Humans , Male , Mice , Models, Biological , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Signal Transduction , Spleen/cytology , Spleen/metabolism
8.
Chest ; 143(2): 461-470, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-22878868

ABSTRACT

BACKGROUND: New evidence links nicotine to the regulation of T cell-mediated inflammation via a 7 nicotinic cholinergic receptor activation, and chronic nicotine exposure (smoking) reduces the incidence of granulomatous diseases. We sought to determine whether nicotine treatment was well tolerated while effectively normalizing immune responses in patients with active pulmonary sarcoidosis. METHODS: Consenting adults with symptomatic sarcoidosis (n 5 13) were randomly assigned to receive 12 weeks of nicotine treatment plus conventional therapy or conventional therapy alone. Obtained blood cells were evaluated for their responsiveness to selected Toll-like receptor (TLR) and nucleotide oligomerization domain-like receptor ligands and T cell surface marker expression before and after nicotine treatment. Asymptomatic patients (n 5 6) and disease-free subjects (n 5 6) served as comparative control subjects. Adverse events were monitored for the duration of the study. RESULTS: Compared with the asymptomatic group, symptomatic patients had impaired peripheral responses to TLR2, TLR4, and TLR9 ligands (anergy) and reduced peripheral populations of CD4 1 FoxP3 1 regulatory T cells (Tregs). Nicotine treatment was associated with restoration of TLR2 and TLR9 responsiveness, and expansion of Tregs, including the CD4 1 CD25 2 FoxP3 1 phenotype. There were no serious adverse events or signs of nicotine dependency. CONCLUSIONS: Nicotine treatment in active pulmonary sarcoidosis was well tolerated and restored peripheral immune responsiveness to TLR2 and TLR9 agonists and expansion of FoxP3 1 Tregs, including a specific "preactivated" (CD25 2 ) phenotype. The immune phenotype of patients with symptomatic sarcoidosis treated with nicotine closely resembled that of asymptomatic patients, supporting the notion that nicotine treatment may be beneficial in this patient population.


Subject(s)
Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Sarcoidosis, Pulmonary/drug therapy , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 9/metabolism , Antigens, Surface/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Immunity, Cellular/drug effects , Male , Middle Aged , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Phenotype , Sarcoidosis, Pulmonary/metabolism , Sarcoidosis, Pulmonary/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 9/immunology , Treatment Outcome
9.
J Immunol ; 189(1): 433-43, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22675199

ABSTRACT

Plasmacytoid dendritic cells (pDC) are potent APCs known to regulate immune responses to self-Ags, particularly DNA. The mitochondrial fraction of necrotic cells was found to most potently promote human pDC activation, as reflected by type I IFN release, which was dependent upon the presence of mitochondrial DNA and involved TLR9 and receptors for advanced glycation end products. Mitochondrial transcription factor A (TFAM), a highly abundant mitochondrial protein that is functionally and structurally homologous to high mobility group box protein 1, was observed to synergize with CpG-containing oligonucleotide, type A, DNA to promote human pDC activation. pDC type I IFN responses to TFAM and CpG-containing oligonucleotide, type A, DNA indicated their engagement with receptors for advanced glycation end products and TLR9, respectively, and were dependent upon endosomal processing and PI3K, ERK, and NF-κB signaling. Taken together, these results indicate that pDC contribute to sterile immune responses by recognizing the mitochondrial component of necrotic cells and further incriminate TFAM and mitochondrial DNA as likely mediators of pDC activation under these circumstances.


Subject(s)
Adjuvants, Immunologic/physiology , CpG Islands/immunology , DNA, Mitochondrial/genetics , DNA-Binding Proteins/physiology , Dendritic Cells/immunology , Dendritic Cells/pathology , Mitochondrial Proteins/physiology , Signal Transduction/immunology , Transcription Factors/physiology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Animals , CpG Islands/genetics , DNA-Binding Proteins/genetics , Dendritic Cells/metabolism , Gene Amplification/immunology , Hep G2 Cells , Humans , Interferon-alpha/metabolism , Mice , Mitochondrial Proteins/genetics , Necrosis , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Toll-Like Receptor 9/physiology , Transcription Factors/genetics
10.
Biochem Biophys Res Commun ; 417(2): 886-91, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22209793

ABSTRACT

BACKGROUND: Recent studies show that various inflammatory diseases are regulated at the level of RNA translation by small non-coding RNAs, termed microRNAs (miRNAs). We sought to determine whether sarcoidosis tissues harbor a distinct pattern of miRNA expression and then considered their potential molecular targets. METHODS AND RESULTS: Genome-wide microarray analysis of miRNA expression in lung tissue and peripheral blood mononuclear cells (PBMCs) was performed and differentially expressed (DE)-miRNAs were then validated by real-time PCR. A distinct pattern of DE-miRNA expression was identified in both lung tissue and PBMCs of sarcoidosis patients. A subgroup of DE-miRNAs common to lung and lymph node tissues were predicted to target transforming growth factor (TGFß)-regulated pathways. Likewise, the DE-miRNAs identified in PBMCs of sarcoidosis patients were predicted to target the TGFß-regulated "wingless and integrase-1" (WNT) pathway. CONCLUSIONS: This study is the first to profile miRNAs in sarcoidosis tissues and to consider their possible roles in disease pathogenesis. Our results suggest that miRNA regulate TGFß and related WNT pathways in sarcoidosis tissues, pathways previously incriminated in the pathogenesis of sarcoidosis.


Subject(s)
Gene Expression Profiling , Integrases/genetics , MicroRNAs/genetics , Molecular Targeted Therapy , Sarcoidosis, Pulmonary/genetics , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics , Gene Expression , Humans , Lung/metabolism , Lymph Nodes/metabolism , Sarcoidosis, Pulmonary/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Wnt Proteins/antagonists & inhibitors
11.
Int J Exp Pathol ; 92(5): 366-76, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21838744

ABSTRACT

Mitochondrial morphology and function are altered in intestinal epithelia during endotoxemia. However, it is unclear whether mitochondrial abnormalities occur in lung epithelial cells during acute sepsis or whether mitochondrial dysfunction corresponds with altered epithelial barrier function. Thus, we hypothesized that the intestinal epithelium is more susceptible to mitochondrial injury than the lung epithelium during acute sepsis and that mitochondrial dysfunction precedes impaired barrier function. Using a resuscitated feline model of Escherichia coli-induced sepsis, lung and ileal tissues were harvested after 6 h for histological and mitochondrial ultrastructural analyses in septic (n = 6) and time-matched controls (n = 6). Human lung epithelial cells (HLEC) and Caco-2 monolayers (n = 5) were exposed to 'cytomix' (TNFα: 40 ng/ml, IL-1ß: 20 ng/ml, IFNγ: 10 ng/ml) for 24-72 h, and measurements of transepithelial electrical resistance (TER), epithelial permeability and mitochondrial membrane potential (ΔΨ) were taken. Lung epithelial morphology, mitochondrial ultrastructure and pulmonary gas exchange were unaltered in septic animals compared to matching controls. While histologically intact, ileal epithelia demonstrated marked mitochondrial ultrastructural damage during sepsis. Caco-2 monolayers treated with cytomix showed a significant decrease in mitochondrial ΔΨ within 24 h, which was associated with a progressive reduction in TER and increased epithelial permeability over the subsequent 48 h. In contrast, mitochondrial ΔΨ and epithelial barrier functions were preserved in HLEC following cytomix. These findings indicate that intestinal epithelium is more susceptible to mitochondrial damage and dysfunction than the lung epithelium in the context of sepsis. Early alterations in mitochondrial function portend subsequent epithelial barrier dysfunction.


Subject(s)
Cell Membrane Permeability/physiology , Escherichia coli Infections/physiopathology , Escherichia coli/isolation & purification , Intestinal Mucosa/physiopathology , Respiratory Mucosa/physiopathology , Sepsis/physiopathology , Acute Disease , Animals , Apoptosis/physiology , Caco-2 Cells , Cats , Cells, Cultured , Colon/microbiology , Colon/pathology , Colon/physiopathology , Disease Models, Animal , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Ileum/microbiology , Ileum/pathology , Ileum/physiopathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lung/microbiology , Lung/pathology , Lung/physiopathology , Male , Membrane Potential, Mitochondrial/physiology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Sepsis/microbiology , Sepsis/pathology , Tumor Necrosis Factor-alpha/blood
12.
Am J Respir Cell Mol Biol ; 44(3): 264-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20889801

ABSTRACT

Mitochondrial antigens released from damaged cells act as "danger signals" capable of promoting innate immune cell migration and activation via formyl peptide receptors (FPRs). Lung epithelial cells are equipped to migrate and mount innate immune responses in the context of acute lung injury. The goal of this study was to determine whether lung epithelial cells express FPRs, which are capable of responding to mitochondrial antigens to promote wound closure and inflammation. Using human Beas2B lung epithelial cells grown to confluency and subjected to linear scratch injury, it was found that mitochondrial antigens enhanced epithelial wound closure, and this phenomenon was inhibited by cyclosporin H, a selective inhibitor of FPR. Although mitochondrial antigens also promoted IL-8 release, this release was not FPR dependent and was unrelated to FPR-induced lung epithelial cell wound closure. The expression of functional FPR was confirmed in Beas2B and primary human tracheobronchial epithelial cells, particularly in lamellipodia at the leading edge of the closing wound. The expression of FPR was increased in response to TNF-α, LPS, scratch injury, and mitochondrial antigen treatment. Considered together, these data confirm that human lung epithelial cells express functional FPRs, which are capable of responding to endogenous mitochondrial danger signals, to promote wound closure.


Subject(s)
Epithelial Cells/cytology , Lung/pathology , Receptors, Formyl Peptide/metabolism , Antigens/chemistry , Cell Line , Cell Movement , Humans , Interleukin-8/metabolism , Ligands , Microscopy, Fluorescence/methods , Mitochondria/pathology , Necrosis/pathology , Subcellular Fractions/metabolism , Wound Healing
13.
Crit Care Med ; 37(6): 2000-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19384205

ABSTRACT

OBJECTIVE: Necrotic cells evoke potent innate immune responses through unclear mechanisms. The mitochondrial fraction of the cell retains constituents of its bacterial ancestors, including N-formyl peptides, which are potentially immunogenic. Thus, we hypothesized that the mitochondrial fraction of the cell, particularly N-formyl peptides, contributes significantly to the activation of monocytes by necrotic cells. DESIGN: Human peripheral blood monocytes were incubated with necrotic cell fractions and mitochondrial proteins to investigate their potential for immune cell activation. SETTING: University Medical Center Research Laboratory. SUBJECTS: Healthy human adults served as blood donors. MEASUREMENTS AND MAIN RESULTS: Human blood monocyte activation was measured after treatment with cytosolic, nuclear and mitochondrial fractions of necrotic HepG2 cells or necrotic HepG2 cells depleted of N-formyl peptides [Rho(0) cells]. The specific role of the high affinity formyl peptide receptor (FPR) was then tested using specific pharmacologic inhibitors and RNA silencing. The capacity of mitochondrial N-formyl peptides to activate monocytes was confirmed using a synthetic peptide conforming to the N-terminus of mitochondrial nicotinamide adenine dinucleotide subunit 6. The results demonstrated that mitochondrial cell fractions most potently activated monocytes, and interleukin (IL)-8 was selectively released at low-protein concentrations. Mitochondria from Rho(0) cells induced minimal monocyte IL-8 release, and specific pharmacologic inhibitors and RNA-silencing confirmed that FPR contributes significantly to monocyte IL-8 responses to both necrotic cells and mitochondrial proteins. N-formyl peptides alone did not induce monocyte IL-8 release; whereas, the combination of mitochondrial N-formyl peptides and mitochondrial transcription factor A (TFAM) dramatically increased IL-8 release from monocytes. Likewise, high mobility group box 1, the nuclear homolog of TFAM, did not induce monocyte IL-8 release unless combined with mitochondrial N-formyl peptides. CONCLUSIONS: Interactions between mitochondrial N-formyl peptides and FPR in the presence of other mitochondrial antigens (e.g., TFAM) contributes significantly to the activation of monocytes by necrotic cells.


Subject(s)
Mitochondrial Proteins/physiology , Monocytes/physiology , Necrosis/pathology , Receptors, Formyl Peptide/physiology , Cells, Cultured , DNA, Mitochondrial/physiology , DNA-Binding Proteins/physiology , Humans , Transcription Factors/physiology
14.
Am J Respir Crit Care Med ; 179(10): 929-38, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19218196

ABSTRACT

RATIONALE: Little is known about the genetic regulation of granulomatous inflammation in sarcoidosis. OBJECTIVES: To determine if tissue gene array analysis would identify novel genes engaged in inflammation and lung remodeling in patients with sarcoidosis. METHODS: Gene expression analysis was performed on tissues obtained from patients with sarcoidosis at the time of diagnosis (untreated) (n = 6) compared with normal lung tissue (n = 6). Expression of select genes was further confirmed in lung tissue from a second series of patients with sarcoidosis and disease-free control subjects (n = 11 per group) by semi-quantitative RT-PCR. Interactive gene networks were identified in patients with sarcoidosis using Ingenuity Pathway Analysis (Ingenuity Systems, Inc., Redwood, CA) software. The expression of proteins corresponding to selected overexpressed genes was determined using fluorokine multiplex analysis, and immunohistochemistry. Selected genes and proteins were then analyzed in bronchoalveolar lavage fluid in an independent series of patients with sarcoidosis (n = 36) and control subjects (n = 12). MEASUREMENTS AND MAIN RESULTS: A gene network engaged in Th1-type responses was most significantly overexpressed in the sarcoidosis lung tissues, including genes not previously reported in the context of sarcoidosis (e.g., IL-7). MMP-12 and ADAMDEC1 transcripts were most highly expressed (> 25-fold) in sarcoidosis lung tissues, corresponding with increased protein expression by immunohistochemistry. MMP-12 and ADAMDEC1 gene and protein expression were increased in bronchoalveolar lavage samples from patients with sarcoidosis, correlating with disease severity. CONCLUSIONS: Tissue gene expression analyses provide novel insights into the pathogenesis of pulmonary sarcoidosis. MMP-12 and ADAMDEC1 emerge as likely mediators of lung damage and/or remodeling and may serve as markers of disease activity.


Subject(s)
Matrix Metalloproteinase 12/genetics , Metalloendopeptidases/genetics , Sarcoidosis, Pulmonary/genetics , ADAM Proteins , Adult , Age Factors , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Granuloma/enzymology , Granuloma/genetics , Humans , Male , Matrix Metalloproteinase 12/biosynthesis , Metalloendopeptidases/biosynthesis , Microarray Analysis , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sarcoidosis, Pulmonary/enzymology , Sarcoidosis, Pulmonary/immunology , Sarcoidosis, Pulmonary/pathology , Sex Factors , Th1 Cells/immunology
15.
Am J Physiol Gastrointest Liver Physiol ; 292(2): G475-81, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17290013

ABSTRACT

In the mammalian small intestine, coupled NaCl absorption occurs via the dual operation of Na/H and Cl/HCO(3) exchange on the villus cell brush border membrane (BBM). Although constitutive nitric oxide (cNO) has been demonstrated to alter gastrointestinal tract functions, how cNO may specifically alter these two transporters to regulate coupled NaCl absorption is unknown. In villus cells, inhibition of cNO synthase (cNOS) with l-N(G)-nitroarginine methylester (l-NAME) stimulated Na/H exchange whereas Cl/HCO(3) exchange was unaffected. In villus cell BBM vesicles (BBMV) prepared from rabbits treated with l-NAME, Na/H exchange was also stimulated. d-NAME, an inactive analog of l-NAME, and N(6)-(1-imonoethyl)-l-lysine dihydrochloride, a more selective inhibitor of inducible NO synthase, did not affect Na/H exchange. Kinetic studies demonstrated that the mechanism of stimulation is secondary to an increase in the maximal rate of uptake of Na, without an alteration in the affinity of the transporter for Na. Northern blot studies demonstrated an increase in the message for the BBM Na/H exchanger NHE3, and Western blot studies showed that the immunoreactive protein levels of NHE3 was increased when cNOS was inhibited. Thus these results indicate that cNO under nominal physiological states most likely maintains an inhibitory tone on small intestinal coupled NaCl absorption by specifically inhibiting BBM Na/H expression.


Subject(s)
Intestinal Mucosa/metabolism , Microvilli/metabolism , Nitric Oxide/metabolism , Sodium-Hydrogen Exchangers/metabolism , Ammonium Chloride/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Enteritis/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Hydrogen-Ion Concentration/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Kinetics , Lysine/analogs & derivatives , Lysine/pharmacology , Microvilli/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Rabbits , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
16.
J Pediatr Gastroenterol Nutr ; 43(2): 168-79, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877980

ABSTRACT

OBJECTIVES: The transfer of exogenous genetic material to cells within the gastrointestinal (GI) tract has many potential therapeutic applications. An attractive feature of the GI tract for gene transfer is its accessibility through the orogastric route. In this study, we evaluated the stability of recombinant adeno-associated virus type 2 (rAAV2) vectors within the GI tract and whether rAAV2-mediated gene transfer could be increased through manipulation of the intraluminal environment. METHODS: The stability of rAAV2 vectors carrying beta-galactosidase and enhanced green fluorescence protein transgenes was determined in the presence of hydrochloric acid, pepsin, trypsin, chymotrypsin gastric fluid and intestinal fluid and after in vivo administration. For in vivo experiments, the rAAV2 vector carrying the beta-galactosidase transgene was administered perorally to FVB/NJ mice. Groups of mice received the vector alone or in combination with sodium bicarbonate and aprotinin. Gene transfer to the stomach and small intestine was evaluated by polymerase chain reaction and histochemical assays. RESULTS: The stability of rAAV2 was reduced by hydrochloric acid, trypsin, chymotrypsin, gastric fluid and intestinal fluid. The vector was not stable within the lumen of the GI tract. Gastric acid neutralization with sodium bicarbonate and protease inhibition with aprotinin increased the in vivo stability of the vector and the level of gene transfer to the stomach and all regions of the small bowel. In both groups of mice (vector alone and vector plus sodium bicarbonate and aprotinin), transgene-derived protein expression (beta-galactosidase) was below the level of detection of the histochemical assay. CONCLUSIONS: Recombinant AAV2 are adversely affected by physiological conditions within the proximal GI tract. Gastric acid neutralization and inhibition of intestinal protease activity improved rAAV2 stability and increased the level of gene transfer within the GI tract. Despite these changes, transduction of the GI tract after peroral rAAV2 administration remained low.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , beta-Galactosidase/metabolism , Animals , Aprotinin/pharmacology , Biotechnology , Dose-Response Relationship, Drug , Gastric Acid/metabolism , Genetic Engineering , Genetic Therapy/methods , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Intestinal Mucosa/metabolism , Mice , Mice, Inbred Strains , Polymerase Chain Reaction , Sodium Bicarbonate/pharmacology , Specific Pathogen-Free Organisms , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics
17.
Am J Physiol Gastrointest Liver Physiol ; 289(6): G1030-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16099871

ABSTRACT

Na-nutrient cotransport processes are not only important for the assimilation of essential nutrients but also for the absorption of Na in the mammalian small intestine. The effect of constitutive nitric oxide (cNO) on Na-glucose (SGLT-1) and Na-amino acid cotransport (NAcT) in the mammalian small intestine is unknown. Inhibition of cNO synthase with N(G)-nitro-l-arginine methyl ester (L-NAME) resulted in the inhibition of Na-stimulated (3)H-O-methyl-D-glucose uptake in villus cells. However, Na-stimulated alanine uptake was not affected in these cells. The L-NAME-induced reduction in SGLT-1 in villus cells was not secondary to an alteration in basolateral membrane Na-K-ATPase activity, which provides the favorable Na gradient for this cotransport process. In fact, SGLT-1 was inhibited in villus cell brush-border membrane (BBM) vesicles prepared from animals treated with L-NAME. Kinetic studies demonstrated that the mechanism of inhibition of SGLT-1 was secondary to a decrease in the affinity for glucose without a change in the maximal rate of uptake of glucose. Northern blot studies demonstrated no change in the mRNA levels of SGLT-1. Western blot studies demonstrated no significant change in the immunoreactive protein levels of SGLT-1 in ileal villus cell BBM from L-NAME-treated rabbits. These studies indicate that inhibition of cNO production inhibits SGLT-1 but not NAcT in the rabbit small intestine. Therefore, whereas cNO promotes Na-glucose cotransport, it does not affect NAcT in the mammalian small intestine.


Subject(s)
Amino Acid Transport Systems, Neutral/physiology , Intestinal Absorption/drug effects , Intestine, Small/physiology , Nitric Oxide/physiology , Animals , Intestine, Small/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Rabbits , Sodium-Glucose Transporter 1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL