Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Am J Pathol ; 194(6): 1090-1105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38403162

ABSTRACT

Changes in the anterior segment of the eye due to type 2 diabetes mellitus (T2DM) are not well-characterized, in part due to the lack of a reliable animal model. This study evaluated changes in the anterior segment, including crystalline lens health, corneal endothelial cell density, aqueous humor metabolites, and ciliary body vasculature, in a rat model of T2DM compared with human eyes. Male Sprague-Dawley rats were fed a high-fat diet (45% fat) or normal diet, and rats fed the high-fat diet were injected with streptozotocin intraperitoneally to generate a model of T2DM. Cataract formation and corneal endothelial cell density were assessed using microscopic analysis. Diabetes-related rat aqueous humor alterations were assessed using metabolomics screening. Transmission electron microscopy was used to assess qualitative ultrastructural changes ciliary process microvessels at the site of aqueous formation in the eyes of diabetic rats and humans. Eyes from the diabetic rats demonstrated cataracts, lower corneal endothelial cell densities, altered aqueous metabolites, and ciliary body ultrastructural changes, including vascular endothelial cell activation, pericyte degeneration, perivascular edema, and basement membrane reduplication. These findings recapitulated diabetic changes in human eyes. These results support the use of this model for studying ocular manifestations of T2DM and support a hypothesis postulating blood-aqueous barrier breakdown and vascular leakage at the ciliary body as a mechanism for diabetic anterior segment pathology.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Rats, Sprague-Dawley , Animals , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Male , Rats , Humans , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Anterior Eye Segment/pathology , Aqueous Humor/metabolism , Cataract/pathology , Cataract/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/metabolism , Lens, Crystalline/ultrastructure , Ciliary Body/pathology , Ciliary Body/metabolism , Diet, High-Fat/adverse effects
4.
Mol Cell Biol ; 42(10): e0016322, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36125265

ABSTRACT

Insulin and insulin-like growth factor 1 (IGF1) signaling is transduced by insulin receptor substrate 1 (IRS1) and IRS2. To elucidate physiological and redundant roles of insulin and IGF1 signaling in adult hearts, we generated mice with inducible cardiomyocyte-specific deletion of insulin and IGF1 receptors or IRS1 and IRS2. Both models developed dilated cardiomyopathy, and most mice died by 8 weeks post-gene deletion. Heart failure was characterized by cardiomyocyte loss and disarray, increased proapoptotic signaling, and increased autophagy. Suppression of autophagy by activating mTOR signaling did not prevent heart failure. Transcriptional profiling revealed reduced serum response factor (SRF) transcriptional activity and decreased mRNA levels of genes encoding sarcomere and gap junction proteins as early as 3 days post-gene deletion, in concert with ultrastructural evidence of sarcomere disruption and intercalated discs within 1 week after gene deletion. These data confirm conserved roles for constitutive insulin and IGF1 signaling in suppressing autophagic and apoptotic signaling in the adult heart. The present study also identifies an unexpected role for insulin and IGF1 signaling in regulating an SRF-mediated transcriptional program, which maintains expression of genes encoding proteins that support sarcomere integrity in the adult heart, reduction of which results in rapid development of heart failure.


Subject(s)
Heart Failure , Insulin-Like Growth Factor I , Mice , Animals , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Insulin/metabolism , Serum Response Factor/metabolism , Sarcomeres/metabolism , Myocytes, Cardiac/metabolism , Heart Failure/metabolism , TOR Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Connexins/metabolism
5.
J Infect Dis ; 223(5): 785-795, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33277988

ABSTRACT

Studies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients and experimentally infected animals indicate a critical role for augmented expression of proinflammatory chemokines and cytokines in severe disease. Here, we demonstrate that SARS-CoV-2 infection of human monocyte-derived macrophages (MDMs) and monocyte-derived dendritic cells was abortive, but induced the production of multiple antiviral and proinflammatory cytokines (interferon-α, interferon-ß, tumor necrosis factor, and interleukins 1ß, 6, and 10) and a chemokine (CXCL10). Despite the lack of efficient replication in MDMs, SARS-CoV-2 induced profound interferon-mediated cell death of host cells. Macrophage activation and death were not enhanced by exposure to low levels of convalescent plasma, suggesting that antibody-dependent enhancement of infection does not contribute to cell death. Together, these results indicate that infection of macrophages and dendritic cells potentially plays a major role in coronavirus disease 2019 pathogenesis, even in the absence of productive infection.


Subject(s)
COVID-19/therapy , Dendritic Cells/virology , Macrophages/virology , SARS-CoV-2/immunology , COVID-19/immunology , Cell Death , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/ultrastructure , Humans , Immunization, Passive , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Macrophages/immunology , Macrophages/ultrastructure , Microscopy, Electron, Transmission , RNA, Messenger/metabolism , RNA, Viral/metabolism , COVID-19 Serotherapy
6.
Arterioscler Thromb Vasc Biol ; 38(6): 1333-1345, 2018 06.
Article in English | MEDLINE | ID: mdl-29599132

ABSTRACT

OBJECTIVE: The main objective of this study is to define the mechanisms by which mitochondria control vascular smooth muscle cell (VSMC) migration and impact neointimal hyperplasia. APPROACH AND RESULTS: The multifunctional CaMKII (Ca2+/calmodulin-dependent kinase II) in the mitochondrial matrix of VSMC drove a feed-forward circuit with the mitochondrial Ca2+ uniporter (MCU) to promote matrix Ca2+ influx. MCU was necessary for the activation of mitochondrial CaMKII (mtCaMKII), whereas mtCaMKII phosphorylated MCU at the regulatory site S92 that promotes Ca2+ entry. mtCaMKII was necessary and sufficient for platelet-derived growth factor-induced mitochondrial Ca2+ uptake. This effect was dependent on MCU. mtCaMKII and MCU inhibition abrogated VSMC migration and mitochondrial translocation to the leading edge. Overexpression of wild-type MCU, but not MCU S92A, mutant in MCU-/- VSMC rescued migration and mitochondrial mobility. Inhibition of microtubule, but not of actin assembly, blocked mitochondrial mobility. The outer mitochondrial membrane GTPase Miro-1 promotes mitochondrial mobility via microtubule transport but arrests it in subcellular domains of high Ca2+ concentrations. In Miro-1-/- VSMC, mitochondrial mobility and VSMC migration were abolished, and overexpression of mtCaMKII or a CaMKII inhibitory peptide in mitochondria (mtCaMKIIN) had no effect. Consistently, inhibition of mtCaMKII increased and prolonged cytosolic Ca2+ transients. mtCaMKII inhibition diminished phosphorylation of focal adhesion kinase and myosin light chain, leading to reduced focal adhesion turnover and cytoskeletal remodeling. In a transgenic model of selective mitochondrial CaMKII inhibition in VSMC, neointimal hyperplasia was significantly reduced after vascular injury. CONCLUSIONS: These findings identify mitochondrial CaMKII as a key regulator of mitochondrial Ca2+ uptake via MCU, thereby controlling mitochondrial translocation and VSMC migration after vascular injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Carotid Artery Injuries/enzymology , Cell Movement , Mitochondria, Muscle/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neointima , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Disease Models, Animal , Hyperplasia , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
7.
PLoS One ; 12(10): e0186311, 2017.
Article in English | MEDLINE | ID: mdl-29059213

ABSTRACT

The multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a serine/threonine kinase important in transducing intracellular Ca2+ signals. While in vitro data regarding the role of CaMKII in the regulation of endothelial nitric oxide synthase (eNOS) are contradictory, its role in endothelial function in vivo remains unknown. Using two novel transgenic models to express CaMKII inhibitor peptides selectively in endothelium, we examined the effect of CaMKII on eNOS activation, NO production, vasomotor tone and blood pressure. Under baseline conditions, CaMKII activation was low in the aortic wall. Consistently, systolic and diastolic blood pressure, heart rate and plasma NO levels were unaltered by endothelial CaMKII inhibition. Moreover, endothelial CaMKII inhibition had no significant effect on NO-dependent vasodilation. These results were confirmed in studies of aortic rings transduced with adenovirus expressing a CaMKII inhibitor peptide. In cultured endothelial cells, bradykinin treatment produced the anticipated rapid influx of Ca2+ and transient CaMKII and eNOS activation, whereas CaMKII inhibition blocked eNOS phosphorylation on Ser-1179 and dephosphorylation at Thr-497. Ca2+/CaM binding to eNOS and resultant NO production in vitro were decreased under CaMKII inhibition. Our results demonstrate that CaMKII plays an important role in transient bradykinin-driven eNOS activation in vitro, but does not regulate NO production, vasorelaxation or blood pressure in vivo under baseline conditions.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Animals , Cell Line , Humans , Phosphorylation
8.
EBioMedicine ; 18: 41-49, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28229908

ABSTRACT

High-dose chemotherapies to treat multiple myeloma (MM) can be life-threatening due to toxicities to normal cells and there is a need to target only tumor cells and/or lower standard drug dosage without losing efficacy. We show that pharmacologically-dosed ascorbic acid (PAA), in the presence of iron, leads to the formation of highly reactive oxygen species (ROS) resulting in cell death. PAA selectively kills CD138+ MM tumor cells derived from MM and smoldering MM (SMM) but not from monoclonal gammopathy undetermined significance (MGUS) patients. PAA alone or in combination with melphalan inhibits tumor formation in MM xenograft mice. This study shows PAA efficacy on primary cancer cells and cell lines in vitro and in vivo.


Subject(s)
Apoptosis/drug effects , Ascorbic Acid/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Ascorbic Acid/chemistry , Ascorbic Acid/therapeutic use , Calcium-Binding Proteins , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drug Therapy, Combination , Humans , Iron/chemistry , Melphalan/therapeutic use , Mice , Mice, Inbred NOD , Microfilament Proteins , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Reactive Oxygen Species/metabolism , Syndecan-1/metabolism , Transplantation, Heterologous
9.
Am J Physiol Lung Cell Mol Physiol ; 310(1): L86-94, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26545899

ABSTRACT

The calcium and calmodulin-dependent kinase II (CaMKII) translates increases in intracellular Ca(2+) into downstream signaling events. Its function in pulmonary pathologies remains largely unknown. CaMKII is a well-known mediator of apoptosis and regulator of endoplasmic reticulum (ER) Ca(2+). ER stress and apoptosis of type II pneumocytes lead to aberrant tissue repair and progressive collagen deposition in pulmonary fibrosis. Thus we hypothesized that CaMKII inhibition alleviates fibrosis in response to bleomycin by attenuating apoptosis and ER stress of type II pneumocytes. We first established that CaMKII was strongly expressed in the distal respiratory epithelium, in particular in surfactant protein-C-positive type II pneumocytes, and activated after bleomycin instillation. We generated a novel transgenic model of inducible expression of the CaMKII inhibitor peptide AC3-I limited to type II pneumocytes (Tg SPC-AC3-I). Tg SPC-AC3-I mice were protected from development of pulmonary fibrosis after bleomycin exposure compared with wild-type mice. CaMKII inhibition also provided protection from apoptosis in type II pneumocytes in vitro and in vivo. Moreover, intracellular Ca(2+) levels and ER stress were increased by bleomycin and significantly blunted with CaMKII inhibition in vitro. These data demonstrate that CaMKII inhibition prevents type II pneumocyte apoptosis and development of pulmonary fibrosis in response to bleomycin. CaMKII inhibition may therefore be a promising approach to prevent or ameliorate the progression of pulmonary fibrosis.


Subject(s)
Alveolar Epithelial Cells/drug effects , Apoptosis/drug effects , Bleomycin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium/metabolism , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Alveolar Epithelial Cells/metabolism , Animals , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Mice, Transgenic , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
10.
J Control Release ; 218: 22-8, 2015 Nov 28.
Article in English | MEDLINE | ID: mdl-26415855

ABSTRACT

There exists a dire need for improved therapeutics to achieve predictable bone regeneration. Gene therapy using non-viral vectors that are safe and efficient at transfecting target cells is a promising approach to overcoming the drawbacks of protein delivery of growth factors. Here, we investigated the transfection efficiency, cytotoxicity, osteogenic potential and in vivo bone regenerative capacity of chemically modified ribonucleic acid (cmRNA) (encoding BMP-2) complexed with polyethylenimine (PEI) and made comparisons with PEI complexed with conventional plasmid DNA (encoding BMP-2). The polyplexes were fabricated at an amine (N) to phosphate (P) ratio of 10 and characterized for transfection efficiency using human bone marrow stromal cells (BMSCs). The osteogenic potential of BMSCs treated with these polyplexes was validated by determining the expression of bone-specific genes, osteocalcin and alkaline phosphatase as well as through the detection of bone matrix deposition. Using a calvarial bone defect model in rats, it was shown that PEI-cmRNA (encoding BMP-2)-activated matrices promoted significantly enhanced bone regeneration compared to PEI-plasmid DNA (BMP-2)-activated matrices. Our proof of concept study suggests that scaffolds loaded with non-viral vectors harboring cmRNA encoding osteogenic proteins may be a powerful tool for stimulating bone regeneration with significant potential for clinical translation.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration , Polyethyleneimine/chemistry , RNA/administration & dosage , RNA/chemistry , Alkaline Phosphatase/genetics , Animals , Bone Marrow Cells/cytology , Cell Survival , Craniocerebral Trauma/therapy , DNA/administration & dosage , DNA/chemistry , Genetic Therapy , Humans , Male , Mice, Inbred BALB C , Osteocalcin/genetics , Plasmids , RNA/pharmacology , RNA/therapeutic use , Rats, Inbred F344 , Stromal Cells/metabolism
11.
Proc Natl Acad Sci U S A ; 112(32): E4400-9, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26216965

ABSTRACT

Compartmentalization and polarized protein trafficking are essential for many cellular functions. The photoreceptor outer segment (OS) is a sensory compartment specialized for phototransduction, and it shares many features with primary cilia. As expected, mutations disrupting protein trafficking to cilia often disrupt protein trafficking to the OS and cause photoreceptor degeneration. Bardet-Biedl syndrome (BBS) is one of the ciliopathies associated with defective ciliary trafficking and photoreceptor degeneration. However, precise roles of BBS proteins in photoreceptor cells and the underlying mechanisms of photoreceptor degeneration in BBS are not well understood. Here, we show that accumulation of non-OS proteins in the OS underlies photoreceptor degeneration in BBS. Using a newly developed BBS mouse model [Leucine zipper transcription factor-like 1 (Lztfl1)/Bbs17 mutant], isolated OSs, and quantitative proteomics, we determined 138 proteins that are enriched more than threefold in BBS mutant OS. In contrast, only eight proteins showed a more than threefold reduction. We found striking accumulation of Stx3 and Stxbp1/Munc18-1 and loss of polarized localization of Prom1 within the Lztfl1 and Bbs1 mutant OS. Ultrastructural analysis revealed that large vesicles are formed in the BBS OS, disrupting the lamellar structure of the OS. Our findings suggest that accumulation (and consequent sequestration) of non-OS proteins in the OS is likely the primary cause of photoreceptor degeneration in BBS. Our data also suggest that a major function of BBS proteins in photoreceptors is to transport proteins from the OS to the cell body or to prevent entry of non-OS proteins into the OS.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , Eye Proteins/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/pathology , Animals , Antibody Specificity , Blotting, Western , Cell Separation , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/complications , Obesity/pathology , Proteomics , Reproducibility of Results , Retinal Degeneration/complications , Retinal Degeneration/physiopathology , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Transcription Factors/metabolism , Ultracentrifugation
12.
Proc Natl Acad Sci U S A ; 112(7): E786-95, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646431

ABSTRACT

Alcohol is the most commonly abused drug worldwide, and chronic alcohol consumption is a major etiological factor in the development of multiple pathological sequelae, including alcoholic cardiomyopathy and hepatic cirrhosis. Here, we identify regulator of G protein signaling 6 (RGS6) as a critical regulator of both alcohol-seeking behaviors and the associated cardiac and hepatic morbidities through two mechanistically divergent signaling actions. RGS6(-/-) mice consume less alcohol when given free access and are less susceptible to alcohol-induced reward and withdrawal. Antagonism of GABA(B) receptors or dopamine D2 receptors partially reversed the reduction in alcohol consumption in RGS6(-/-) animals. Strikingly, dopamine transporter inhibition completely restored alcohol seeking in mice lacking RGS6. RGS6 deficiency was associated with alterations in the expression of genes controlling dopamine (DA) homeostasis and a reduction in DA levels in the striatum. Taken together, these data implicate RGS6 as an essential regulator of DA bioavailability. RGS6 deficiency also provided dramatic protection against cardiac hypertrophy and fibrosis, hepatic steatosis, and gastrointestinal barrier dysfunction and endotoxemia when mice were forced to consume alcohol. Although RGS proteins canonically function as G-protein regulators, RGS6-dependent, alcohol-mediated toxicity in the heart, liver, and gastrointestinal tract involves the ability of RGS6 to promote reactive oxygen species-dependent apoptosis, an action independent of its G-protein regulatory capacity. We propose that inhibition of RGS6 might represent a viable means to reduce alcohol cravings and withdrawal in human patients, while simultaneously protecting the heart and liver from further damage upon relapse.


Subject(s)
Alcohol Drinking , Behavior, Animal , RGS Proteins/physiology , Reward , Animals , Apoptosis/physiology , Cardiomyopathies/etiology , Conditioning, Operant , Mice , Mice, Knockout , RGS Proteins/genetics
13.
PLoS One ; 10(1): e0116582, 2015.
Article in English | MEDLINE | ID: mdl-25629318

ABSTRACT

Type 1 diabetes (T1D) is caused by autoimmune disease that leads to the destruction of pancreatic ß-cells. Transplantation of cadaveric pancreatic organs or pancreatic islets can restore normal physiology. However, there is a chronic shortage of cadaveric organs, limiting the treatment of the majority of patients on the pancreas transplantation waiting list. Here, we hypothesized that human iPS cells can be directly differentiated into insulin producing cells (IPCs) capable of secreting insulin. Using a series of pancreatic growth factors, we successfully generated iPS cells derived IPCs. Furthermore, to investigate the capability of these cells to secrete insulin in vivo, the differentiated cells were transplanted under the kidney capsules of diabetic immunodeficient mice. Serum glucose levels gradually declined to either normal or near normal levels over 150 days, suggesting that the IPCs were secreting insulin. In addition, using MRI, a 3D organoid appeared as a white patch on the transplanted kidneys but not on the control kidneys. These organoids showed neo-vascularization and stained positive for insulin and glucagon. All together, these data show that a pancreatic organ can be created in vivo providing evidence that iPS cells might be a novel option for the treatment of T1D.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Neovascularization, Physiologic , Stem Cell Transplantation , Animals , Blood Glucose , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Organoids , Oxygen Consumption
15.
Biomaterials ; 35(2): 737-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161167

ABSTRACT

Gene therapy using non-viral vectors that are safe and efficient in transfecting target cells is an effective approach to overcome the shortcomings of protein delivery of growth factors. The objective of this study was to develop and test a non-viral gene delivery system for bone regeneration utilizing a collagen scaffold to deliver polyethylenimine (PEI)-plasmid DNA (pDNA) [encoding platelet derived growth factor-B (PDGF-B)] complexes. The PEI-pPDGF-B complexes were fabricated at amine (N) to phosphate (P) ratio of 10 and characterized for size, surface charge, and in vitro cytotoxicity and transfection efficacy in human bone marrow stromal cells (BMSCs). The influence of the complex-loaded collagen scaffold on cellular attachment and recruitment was evaluated in vitro using microscopy techniques. The in vivo regenerative capacity of the gene delivery system was assessed in 5 mm diameter critical-sized calvarial defects in Fisher 344 rats. The complexes were ~100 nm in size with a positive surface charge. Complexes prepared at an N/P ratio of 10 displayed low cytotoxicity as assessed by a cell viability assay. Confocal microscopy revealed significant proliferation of BMSCs on complex-loaded collagen scaffolds compared to empty scaffolds. In vivo studies showed significantly higher new bone volume/total volume (BV/TV) % in calvarial defects treated with the complex-activated scaffolds following 4 weeks of implantation (14- and 44-fold higher) when compared to empty defects or empty scaffolds, respectively. Together, these findings suggest that non-viral PDGF-B gene-activated scaffolds are effective for bone regeneration and are an attractive gene delivery system with significant potential for clinical translation.


Subject(s)
Bone Regeneration/genetics , Gene Transfer Techniques , Platelet-Derived Growth Factor/genetics , Tissue Scaffolds/chemistry , Animals , Cell Proliferation , Cell Survival , Collagen/chemistry , DNA , Gene Expression , Genetic Therapy , Genetic Vectors , Humans , Male , Mesenchymal Stem Cells , Microscopy, Confocal , Plasmids/genetics , Platelet-Derived Growth Factor/metabolism , Polyethyleneimine , Rats , Rats, Inbred F344 , Transfection
16.
Sci Transl Med ; 5(195): 195ra97, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884469

ABSTRACT

Increased reactive oxygen species (ROS) contribute to asthma, but little is known about the molecular mechanisms connecting increased ROS with characteristic features of asthma. We show that enhanced oxidative activation of the Ca(2+)/calmodulin-dependent protein kinase (ox-CaMKII) in bronchial epithelium positively correlates with asthma severity and that epithelial ox-CaMKII increases in response to inhaled allergens in patients. We used mouse models of allergic airway disease induced by ovalbumin (OVA) or Aspergillus fumigatus (Asp) and found that bronchial epithelial ox-CaMKII was required to increase a ROS- and picrotoxin-sensitive Cl(-) current (ICl) and MUC5AC expression, upstream events in asthma progression. Allergen challenge increased epithelial ROS by activating NADPH oxidases. Mice lacking functional NADPH oxidases due to knockout of p47 and mice with epithelial-targeted transgenic expression of a CaMKII inhibitory peptide or wild-type mice treated with inhaled KN-93, an experimental small-molecule CaMKII antagonist, were protected against increases in ICl, MUC5AC expression, and airway hyperreactivity to inhaled methacholine. Our findings support the view that CaMKII is a ROS-responsive, pluripotent proasthmatic signal and provide proof-of-concept evidence that CaMKII is a therapeutic target in asthma.


Subject(s)
Asthma/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Administration, Intranasal , Animals , Asthma/drug therapy , Asthma/metabolism , Benzylamines/administration & dosage , Benzylamines/therapeutic use , Blotting, Western , Bronchi/metabolism , Bronchi/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Female , Humans , In Vitro Techniques , Male , Mice , NADPH Oxidases/metabolism , Ovalbumin/pharmacology , Oxidation-Reduction , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use
17.
Nature ; 491(7423): 269-73, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23051746

ABSTRACT

Myocardial cell death is initiated by excessive mitochondrial Ca(2+) entry causing Ca(2+) overload, mitochondrial permeability transition pore (mPTP) opening and dissipation of the mitochondrial inner membrane potential (ΔΨm). However, the signalling pathways that control mitochondrial Ca(2+) entry through the inner membrane mitochondrial Ca(2+) uniporter (MCU) are not known. The multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is activated in ischaemia reperfusion, myocardial infarction and neurohumoral injury, common causes of myocardial death and heart failure; these findings suggest that CaMKII could couple disease stress to mitochondrial injury. Here we show that CaMKII promotes mPTP opening and myocardial death by increasing MCU current (I(MCU)). Mitochondrial-targeted CaMKII inhibitory protein or cyclosporin A, an mPTP antagonist with clinical efficacy in ischaemia reperfusion injury, equivalently prevent mPTP opening, ΔΨm deterioration and diminish mitochondrial disruption and programmed cell death in response to ischaemia reperfusion injury. Mice with myocardial and mitochondrial-targeted CaMKII inhibition have reduced I(MCU) and are resistant to ischaemia reperfusion injury, myocardial infarction and neurohumoral injury, suggesting that pathological actions of CaMKII are substantially mediated by increasing I(MCU). Our findings identify CaMKII activity as a central mechanism for mitochondrial Ca(2+) entry in myocardial cell death, and indicate that mitochondrial-targeted CaMKII inhibition could prevent or reduce myocardial death and heart failure in response to common experimental forms of pathophysiological stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/enzymology , Myocardium/pathology , Stress, Physiological , Animals , Apoptosis/drug effects , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Cyclosporine/pharmacology , Female , Heart/drug effects , Heart/physiopathology , Heart Failure/drug therapy , Heart Failure/prevention & control , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Infarction/drug therapy , Myocardial Infarction/prevention & control , Myocardium/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Serine/metabolism , Stress, Physiological/drug effects
18.
Am J Surg ; 204(3): 384-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22494881

ABSTRACT

BACKGROUND: A previous study showed that running polypropylene sutures anchored with square knots retain only 75% of their strength compared with half hitches. The aim of this study was to investigate whether anchor knot geometry similarly affects the tensile strength of other types of sutures used in continuous closures. METHODS: Monofilament and multifilament sutures (all 3-0) were anchored with either square knots or half hitches to 1 tensionometer post, and the running ends were secured to the other. The force required to break the running suture and the site of suture failure were recorded. RESULTS: The running sutures anchored with square knots retained only 50% to 84% of the strength of the identical sutures secured with half hitches (P < .001). CONCLUSIONS: A running suture anchored with half hitches is stronger and safer in comparison with the same suture anchored with square knots. This study provokes a fundamental reconsideration of the use of square knots to anchor running sutures.


Subject(s)
Suture Techniques , Sutures , Catgut , Dioxanes , Humans , Microscopy, Electron, Scanning , Nylons , Polydioxanone , Polyesters , Polyglactin 910 , Silk , Tensile Strength
19.
Nat Med ; 17(12): 1610-8, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081025

ABSTRACT

Excessive activation of the ß-adrenergic, angiotensin II (Ang II) and aldosterone signaling pathways promotes mortality after myocardial infarction, and antagonists targeting these pathways are core therapies for treating this condition. Catecholamines and Ang II activate the multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), the inhibition of which prevents isoproterenol-mediated and Ang II-mediated cardiomyopathy. Here we show that aldosterone exerts direct toxic actions on myocardium by oxidative activation of CaMKII, causing cardiac rupture and increased mortality in mice after myocardial infarction. Aldosterone induces CaMKII oxidation by recruiting NADPH oxidase, and this oxidized and activated CaMKII promotes matrix metalloproteinase 9 (MMP9) expression in cardiomyocytes. Myocardial CaMKII inhibition, overexpression of methionine sulfoxide reductase A (an enzyme that reduces oxidized CaMKII) or NADPH oxidase deficiency prevented aldosterone-enhanced cardiac rupture after myocardial infarction. These findings show that oxidized myocardial CaMKII mediates the cardiotoxic effects of aldosterone on the cardiac matrix and establish CaMKII as a nodal signal for the neurohumoral pathways associated with poor outcomes after myocardial infarction.


Subject(s)
Aldosterone/adverse effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotoxins/adverse effects , Myocardial Infarction/pathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Heart/drug effects , Humans , Luciferases/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Methionine Sulfoxide Reductases/genetics , Methionine Sulfoxide Reductases/metabolism , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Oxidation-Reduction , Signal Transduction , Up-Regulation
20.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L569-78, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21257729

ABSTRACT

During pulmonary edema, the alveolar space is exposed to a hypoxic environment. The integrity of the alveolar epithelial barrier is required for the reabsorption of alveolar fluid. Tight junctions (TJ) maintain the integrity of this barrier. We set out to determine whether hypoxia creates a dysfunctional alveolar epithelial barrier, evidenced by an increase in transepithelial electrical conductance (G(t)), due to a decrease in the abundance of TJ proteins at the plasma membrane. Alveolar epithelial cells (AEC) exposed to mild hypoxia (Po(2) = 50 mmHg) for 30 and 60 min decreased occludin abundance at the plasma membrane and significantly increased G(t). Other cell adhesion molecules such as E-cadherin and claudins were not affected by hypoxia. AEC exposed to hypoxia increased superoxide, but not hydrogen peroxide (H(2)O(2)). Overexpression of superoxide dismutase 1 (SOD1) but not SOD2 prevented the hypoxia-induced G(t) increase and occludin reduction in AEC. Also, overexpression of catalase had a similar effect as SOD1, despite not detecting any increase in H(2)O(2) during hypoxia. Blocking PKC-ζ and protein phosphatase 2A (PP2A) prevented the hypoxia-induced occludin reduction at the plasma membrane and increase in G(t). In summary, we show that superoxide, PKC-ζ, and PP2A are involved in the hypoxia-induced increase in G(t) and occludin reduction at the plasma membrane in AEC.


Subject(s)
Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/enzymology , Cell Membrane/metabolism , Electric Conductivity , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Protein Phosphatase 2/metabolism , Animals , Catalase/metabolism , Cell Hypoxia , Hydrogen-Ion Concentration , Lanthanum/metabolism , Male , Occludin , Partial Pressure , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...