Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 60(4): 965-977, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30884523

ABSTRACT

Purpose: Retinal ischemia, a common cause of several vision-threatening diseases, contributes to the death of retinal neurons, particularly retinal ganglion cells (RGCs). Heat shock transcription factor 1 (HSF1), a stress-responsive protein, has been shown to be important in response to cellular stress stimuli, including ischemia. This study is to investigate whether HSF1 has a role in retinal neuronal injury in a mouse model of retinal ischemia-reperfusion (IR). Methods: IR was induced by inserting an infusion needle into the anterior chamber of the right eye and elevating a saline reservoir connected to the needle to raise the intraocular pressure to 110 mm Hg for 45 minutes. HSF1, Hsp70, molecules in the endoplasmic reticulum (ER) stress branches, tau phosphorylation, inflammatory molecules, and RGC injury were determined by immunohistochemistry, Western blot, or quantitative PCR. Results: HSF1 expression was significantly increased in the retina 6 hours after IR. Using our novel transgenic mice carrying full-length human HSF gene, we demonstrated that IR-induced retinal neuronal apoptosis and necroptosis were abrogated 12 hours after IR. RGCs and their function were preserved in the HSF1 transgenic mice 7 days after IR. Mechanistically, the beneficial effects of HSF1 may be mediated by its induction of chaperone protein Hsp70 and alleviation of ER stress, leading to decreased tau phosphorylation and attenuated inflammatory response 12 to 24 hours after IR. Conclusions: These data provide compelling evidence that HSF1 is neuroprotective against retinal IR injury, and boosting HSF1 expression may be a beneficial strategy to limit neuronal degeneration in retinal diseases.


Subject(s)
Gene Expression Regulation/physiology , Heat Shock Transcription Factors/genetics , Optic Nerve Injuries/genetics , Reperfusion Injury/genetics , Retinal Diseases/genetics , Animals , Blotting, Western , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Leukostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Crush , Neuroprotection/physiology , Optic Nerve Injuries/prevention & control , Phosphorylation , Real-Time Polymerase Chain Reaction , Reperfusion Injury/prevention & control , Retinal Diseases/prevention & control , Tomography, Optical Coherence , tau Proteins/metabolism
2.
Invest Ophthalmol Vis Sci ; 59(11): 4670-4682, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30267089

ABSTRACT

Purpose: Retinal ganglion cell (RGC) death following axonal injury occurring in traumatic optic neuropathy (TON) causes irreversible vision loss. GRP78 is a molecular chaperone that enhances protein folding and controls activation of endoplasmic reticulum (ER) stress pathways. This study determined whether adeno-associated virus (AAV)-mediated gene transfer of GRP78 protected RGCs from death in a mouse model of TON induced by optic nerve crush (ONC). Methods: ONC was induced by a transient crush of optic nerve behind the eye globe. AAV was used to deliver genes into retina. Molecules in the ER stress branches, tau oligomers, and RGC injury were determined by immunohistochemistry or Western blot. Results: Among tested AAV serotypes, AAV2 was the most efficient for delivering genes to RGCs. Intravitreal delivery of AAV2-GRP78 markedly attenuated ER stress and RGC death 3 days after ONC, and significantly improved RGC survival and function 7 days after ONC. Protein aggregation is increased during ER stress and aggregated proteins such as tau oligomers are key players in neurodegenerative diseases. AAV2-GRP78 alleviated ONC-induced increases in tau phosphorylation and oligomerization. Furthermore, tau oligomers directly induced RGC death, and blocking tau oligomers with tau oligomer monoclonal antibody (TOMA) attenuated ONC-induced RGC loss. Conclusion: These data indicate that the beneficial effect of AAV2-GRP78 is partially mediated by the reduction of misfolded tau, and provide compelling evidence that gene therapy with AAV2-GRP78 or immunotherapy with TOMA offers novel therapeutic approaches to alleviate RGC loss in TON.


Subject(s)
Dependovirus/genetics , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/genetics , Optic Nerve Injuries/prevention & control , Retinal Ganglion Cells/metabolism , Transfection , tau Proteins/metabolism , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Endoplasmic Reticulum Chaperone BiP , Gene Expression/physiology , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Nerve Crush , Optic Nerve Injuries/metabolism , Protein Aggregates , Reperfusion Injury/prevention & control , Tomography, Optical Coherence
3.
Aging (Albany NY) ; 10(8): 1932-1946, 2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30070973

ABSTRACT

Corneal transparency, dependent on the integrity of epithelial cells, is essential for vision. Corneal epithelial damage is one of the most commonly observed ocular conditions and proper wound healing is necessary for corneal transparency. Sirt6, a histone deacetylase, has been shown to regulate many cellular events including aging and inflammation. However, its specific role in corneal epithelial wound healing remains unknown. Here we demonstrated that Sirt6 was expressed in corneal epithelial cells and its expression decreased with age. In an in vivo corneal epithelial wound healing model, Sirt6 deficiency resulted in delayed and incomplete wound healing and was associated excessive inflammation in the corneal stroma and dysfunction of Notch signaling, leading to keratinization of the corneal epithelium and corneal opacity. Aging Sirt6-deficient mice spontaneously developed corneal keratitis with extensive infiltration of inflammatory cells into the cornea. In vitro experiments demonstrated that primary corneal epithelial cells with Sirt6 downregulation expressed increased basal levels of inflammatory genes and exhibited hyper-inflammatory reactivity to IL-1ß and TNFα treatment. These results provide compelling evidence that Sirt6 is a critical regulator of inflammation in the cornea, and is responsible for corneal epithelial wound healing, thus contributing to the maintenance of epithelial integrity and corneal transparency.


Subject(s)
Aging/physiology , Epithelial Cells/physiology , Epithelium, Corneal/physiology , Sirtuins/metabolism , Wound Healing/physiology , Animals , Cell Movement , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
4.
Arterioscler Thromb Vasc Biol ; 37(10): 1881-1890, 2017 10.
Article in English | MEDLINE | ID: mdl-28818856

ABSTRACT

OBJECTIVE: Infusion of angiotensin II (Ang II) induces extracellular matrix remodeling and inflammation resulting in abdominal aortic aneurysms (AAAs) in normolipidemic mice. Although Ang II activates mesenchymal cells in the media and adventitia to become fibrogenic, the sentinel role of this mesenchymal population in modulating the inflammatory response and aneurysms is not known. We test the hypothesis that these fibrogenic mesenchymal cells play a critical role in Ang II-induced aortic wall vascular inflammation and AAA formation. APPROACH AND RESULTS: Ang II infusion increased phospho-Ser536-RelA and interleukin (IL)-6 immunostaining in the abdominal aorta. In addition, aortic mRNA transcripts of RelA-dependent cytokines IL-6 and IL-1ß were significantly elevated suggesting that Ang II functionally activates RelA signaling. To test the role of mesenchymal RelA in AAA formation, we generated RelA-CKO mice by administering tamoxifen to double transgenic mice harboring RelA-flox alleles and tamoxifen-inducible Col1a2 promoter-driven Cre recombinase (Col1a2-CreERT). Tamoxifen administration to Col1a2-CreERT•mT/mG mice induced Cre expression and RelA depletion in aortic smooth muscle cells and fibroblasts but not in endothelial cells. Infusion of Ang II significantly increased abdominal aortic diameter and the incidence of AAA in RelA wild-type but not in RelA-CKO mice, independent of changes in systolic blood pressure. Furthermore, mesenchymal cell-specific RelA-CKO mice exhibited decreased expression of IL-6 and IL-1ß cytokines and decreased recruitment of C68+ and F4/80lo•Ly6Chi monocytes during Ang II infusion. CONCLUSIONS: Fibrogenic mesenchymal RelA plays a causal role in Ang II-induced vascular inflammation and AAA in normolipidemic mice.


Subject(s)
Aorta, Abdominal/physiopathology , Aorta/physiopathology , Aortic Aneurysm, Abdominal/physiopathology , Mesenchymal Stem Cells/physiology , Transcription Factor RelA/physiology , Angiotensin II/pharmacology , Animals , Aorta/cytology , Blood Pressure/physiology , Collagen Type I/physiology , Integrases/physiology , Mice , Mice, Transgenic , Monocytes/physiology , Tamoxifen/pharmacology
5.
Aging (Albany NY) ; 9(3): 1069-1083, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28351995

ABSTRACT

Aging is associated with an increased incidence and prevalence of renal glomerular diseases. Sirtuin (Sirt) 6, a nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase, has been shown to protect against multiple age-associated phenotypes; however it is unknown whether Sirt6 has a direct pathophysiologic role in the kidney. In the present study, we demonstrate that Sirt6 is expressed in the kidney and aging Sirt6-deficient mice exhibit renal hypertrophy with glomerular enlargement. Sirt6 deletion induces podocyte injury, including decreases in slit diaphragm proteins, foot process effacement, and cellular loss, resulting in proteinuria. Knockdown of Sirt6 in cultured primary murine podocytes induces shape changes with loss of process formation and cell apoptosis. Moreover, Sirt6 deficiency results in progressive renal inflammation and fibrosis. Collectively, these data provide compelling evidence that Sirt6 is important for podocyte homeostasis and maintenance of glomerular function, and warrant further investigation into the role of Sirt6 in age-associated kidney dysfunction.


Subject(s)
Aging/pathology , Kidney Diseases/metabolism , Kidney Glomerulus/pathology , Sirtuins/metabolism , Animals , Disease Progression , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Mice , Mice, Knockout
6.
Cell Death Dis ; 8(2): e2606, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28182006

ABSTRACT

Transdifferentiation of quiescent dermal fibroblasts to secretory myofibroblasts has a central role in wound healing and pathological scar formation. This myofibroblast transdifferentiation process involves TGFß-induced de novo synthesis of alpha smooth muscle cell actin (αSMA)+ fibers that enhance contractility as well as increased expression of extracellular matrix (ECM) proteins, including collagen and fibronectin. These processes are mediated upstream by the reactive oxygen species (ROS)-producing enzyme Nox4, whose induction by TGFß is incompletely understood. In this study, we demonstrate that Nox4 is involved in αSMA+ fiber formation and collagen production in primary human dermal fibroblasts (hDFs) using a small-molecule inhibitor and siRNA-mediated silencing. Furthermore, TGFß-induced signaling via Smad3 is required for myofibroblast transformation and Nox4 upregulation. Immunoprecipitation-selected reaction monitoring (IP-SRM) assays of the activated Smad3 complex suggest that it couples with the epigenetic reader and transcription co-activator bromodomain and extraterminal (BET) domain containing protein 4 (BRD4) to promote Nox4 transcription. In addition, cyclin-dependent kinase 9 (CDK9), a component of positive transcription elongation factor, binds to BRD4 after TGFß stimulation and is also required for RNA polymerase II phosphorylation and Nox4 transcription regulation. Surprisingly, BRD4 depletion decreases myofibroblast differentiation but does not affect collagen or fibronectin expression in primary skin fibroblasts, whereas knockdown of CDK9 decreases all myofibroblast genes. We observe enhanced numbers and persistence of myofibroblast formation and TGFß signaling in hypertrophic scars. BRD4 inhibition reverses hypertrophic skin fibroblast transdifferentiation to myofibroblasts. Our data indicate that BRD4 and CDK9 have independent, coordinated roles in promoting the myofibroblast transition and suggest that inhibition of the Smad3-BRD4 pathway may be a useful strategy to limit hypertrophic scar formation after burn injury.


Subject(s)
Cell Transdifferentiation/physiology , Cyclin-Dependent Kinase 9/metabolism , Myofibroblasts/metabolism , NADPH Oxidases/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Transforming Growth Factor beta/metabolism , Adolescent , Cell Cycle Proteins , Cells, Cultured , Child , Child, Preschool , Collagen/metabolism , Female , Fibronectins/metabolism , Gene Expression Regulation/physiology , Humans , Infant , Infant, Newborn , Male , Myofibroblasts/physiology , NADPH Oxidase 4 , Signal Transduction/physiology , Smad3 Protein , Up-Regulation/physiology , Wound Healing/physiology
7.
Physiol Rep ; 4(21)2016 11.
Article in English | MEDLINE | ID: mdl-27905294

ABSTRACT

Glucocorticoids (GC) are a frontline therapy for numerous acute and chronic diseases because of their demonstrated efficacy at reducing systemic inflammation. An unintended side effect of GC therapy is the stimulation of skeletal muscle atrophy. Pathophysiological mechanisms responsible for GC-induced skeletal muscle atrophy have been extensively investigated, and the ability to treat patients with GC without unintended muscle atrophy has yet to be realized. We have reported that a single, standard-of-care dose of Methylprednisolone increases in vivo expression of NF-κB-inducing kinase (NIK), an important upstream regulatory kinase controlling NF-κB activation, along with other key muscle catabolic regulators such as Atrogin-1 and MuRF1 that induce skeletal muscle proteolysis. Here, we provide experimental evidence that overexpressing NIK by intramuscular injection of recombinant human NIK via adenoviral vector in mouse tibialis anterior muscle induces a 30% decrease in the average fiber cross-sectional area that is associated with increases in mRNA expression of skeletal muscle atrophy biomarkers MuRF1, Atrogin-1, myostatin and Gadd45. A single injection of GC induced NIK mRNA and protein within 2 h, with the increased NIK localized to nuclear and sarcolemmal locations within muscle fibers. Daily GC injections induced skeletal muscle fore limb weakness as early as 3 days with similar atrophy of muscle fibers as observed with NIK overexpression. NIK overexpression in primary human skeletal muscle myotubes increased skeletal muscle atrophy biomarkers, while NIK knockdown significantly attenuated GC-induced increases in NIK and Atrogin-1. These results suggest that NIK may be a novel, previously unrecognized mediator of GC-induced skeletal muscle atrophy.


Subject(s)
Glucocorticoids/pharmacology , Muscle, Skeletal/enzymology , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Glucocorticoids/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/pathology , Protein Serine-Threonine Kinases/administration & dosage , RNA, Messenger/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , NF-kappaB-Inducing Kinase
8.
BMC Dev Biol ; 16(1): 32, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27662828

ABSTRACT

BACKGROUND: Nuclear Factor-Kappa B (NF-kB) is a family of transcription factors that are important in embryonic development, inflammation, epithelial-to-mesenchymal transition and cancer. The 65 kDa RelA subunit is the major transcriptional activator of the NF-kB pathways. Whole-body deficiency of RelA leads to massive apoptosis of liver hepatocytes and death in utero. To study the role of RelA in physiology and in disease states in a manner that circumvents this embryonic lethal phenotype, we have generated a mouse with RelA conditional knockout (CKO) alleles containing loxP sites that are deleted by activated Cre recombinase. RESULTS: We demonstrate that RelACKO/CKO mice are fertile, do not display any developmental defects and can be crossed with Cre-expressing mice to delete RelA in a temporal, tissue-specific manner. Our mating of RelACKO/CKO mice with Zp3-Cre transgenic led to embryonic lethality of RelA-deficient embryos. In contrast, mating of RelACKO/CKO mice with Col1α2-CreER mice allowed for the generation of double transgenics which could be stimulated with tamoxifen to induce fibroblast-specific RelA deletion in adulthood. CONCLUSIONS: Based on our collective data, we conclude that this novel RelACKO/CKO mouse allows for efficient deletion of RelA in a tissue-specific manner. This RelACKO/CKO mouse will be an invaluable tool for deciphering the mechanistic roles of RelA in various cells and tissues during development and in disease.

9.
J Thorac Dis ; 8(8): E746-54, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27619163

ABSTRACT

On April 29, 2015, Son and colleagues published an article entitled "Granulocyte macrophage colony-stimulating factor (GM-CSF) is required for aortic dissection/intramural haematoma" in Nature Communications. The authors observed that the heterozygous Kruppel-like transcription factor 6 (KLF6) deficiency or absence of myeloid-specific KLF6 led to upregulation of macrophage GM-CSF expression, promoted the development of aortic hematoma/dissection, and stimulated abdominal aortic aneurysm (AAA) formation when the vessel wall was subjected to an inflammatory stimulus. The additional findings of increased adventitial fibrotic deposition, marked infiltration of macrophages, and increased expression of matrix metalloprotease-9 (MMP-9) and IL-6 were blocked with neutralizing GM-CSF antibodies, or recapitulated in normal mice with excess GM-CSF administration. The authors concluded that GM-CSF is a key regulatory molecule in the development of AAA and further suggested that activation of GM-CSF is independent of the transforming growth factor ß (TGFß)-Smad pathway associated with the Marfan aortic pathology. In this perspective, we expand on this mechanism, drawing from previous studies implicating a similar essential role for IL-6 signaling in macrophage activation, Th17 expansion and aortic dissections. We propose a sequential "two-hit" model of vascular inflammation involving initial vascular injury followed by recruitment of Ly6C(hi) macrophages. Aided by fibroblast interactions inflammatory macrophages produce amplification of IL-6 and GM-CSF expression that converge on a common, pathogenic Janus kinase (JAK)-signal transducers and activations of transcription 3 (STAT3) signaling pathway. This pathway stimulates effector functions of macrophages, promotes differentiation of Th17 lymphocytes and enhances matrix metalloproteinase expression, ultimately resulting in deterioration of vascular wall structural integrity. Further research evaluating the impact of interventions modulating this common JAK-STAT3 pathway may yield new therapeutic interventions for late stages of vascular expansion in inflammation driven aortic disease.

10.
Free Radic Biol Med ; 79: 176-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25476852

ABSTRACT

The mammalian sirtuin 6 (Sirt6) is a site-specific histone deacetylase that regulates chromatin structure and many fundamental biological processes. It inhibits endothelial cell senescence and inflammation, prevents development of cardiac hypertrophy and heart failure, modulates glucose metabolism, and represses tumor growth. The basic molecular mechanisms underlying regulation of Sirt6 enzymatic function are largely unknown. Here we hypothesized that Sirt6 function can be regulated via posttranslational modification, focusing on the role of peroxynitrite, one of the major reactive nitrogen species formed by excessive nitric oxide and superoxide generated during disease processes. We found that incubation of purified recombinant Sirt6 protein with 3-morpholinosydnonimine (SIN-1; a peroxynitrite donor that generates nitric oxide and superoxide simultaneously) increased Sirt6 tyrosine nitration and decreased its intrinsic catalytic activity. Similar results were observed in SIN-1-treated Sirt6, which was overexpressed in HEK293 cells, and in endogenous Sirt6 when human retinal microvascular endothelial cells were treated with SIN-1. To further investigate whether Sirt6 nitration occurs under pathological conditions, we determined Sirt6 nitration and activity in retina using a model of endotoxin-induced retinal inflammation. Our data showed that Sirt6 nitration was increased, whereas its activity was decreased, in this model. With mass spectrometry, we identified that tyrosine 257 in Sirt6 was nitrated after SIN-1 treatment. Mutation of tyrosine 257 to phenylalanine caused loss of Sirt6 activity and abolished SIN-1-induced nitration and decrease in its activity. Mass spectrometry analysis also revealed oxidation of methionine and tryptophan in Sirt6 after SIN-1 treatment. Our results demonstrate a novel regulatory mechanism controlling Sirt6 activity through reactive nitrogen species-mediated posttranslational modification under oxidative and nitrosative stress.


Subject(s)
Peroxynitrous Acid/pharmacology , Protein Processing, Post-Translational , Sirtuins/metabolism , Amino Acid Sequence , Animals , Chromatography, Liquid , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Oxidation-Reduction , Retina/drug effects , Retina/metabolism , Retina/pathology , Sequence Homology, Amino Acid , Sirtuins/chemistry , Tandem Mass Spectrometry
11.
Curr Eye Res ; 40(4): 368-77, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24911405

ABSTRACT

PURPOSE: To investigate the effects of nicotine on retinal alterations in early-stage diabetes in an established rodent model. MATERIALS AND METHODS: Sprague-Dawley rats were examined using a combination of confocal scanning laser ophthalmoscopy and spectral domain optical coherence tomography to determine changes in retinal structure in response to nicotine exposure, diabetes and the combined effects of nicotine and diabetes. Diabetes was induced by a single injection of 65 mg/kg streptozotocin and nicotine injections were administered subcutaneously daily. Retinal thickness in the superior, inferior, nasal and temporal quadrants were determined based on the spectral domain optical coherence tomography (SD-OCT) volume scans (20° × 20°) centered on the optic disc. Segmentation of discrete retinal layers was performed on a subset of SD-OCT cross-sections to further examine changes in each treatment group. Survival of neurons within the ganglion cell layer (GCL) was assessed by confocal morphometric imaging. RESULTS: The control group did not experience any significant change throughout the study. The nicotine treatment group experienced an average decrease in total retinal thickness (TRT) of 9.4 µm with the majority of the loss localized within the outer nuclear layer (ONL) as determined by segmentation analysis (p < 0.05). The diabetic group exhibited a trend toward decreased TRT while segmentation analysis of the diabetic retinopathy (DR) group revealed significant thinning within the ONL (p < 0.05). The combination of nicotine and diabetes revealed a significant increase of 8.9 µm in the TRT (p < 0.05) accompanied by a decrease in the number of GCL neurons. CONCLUSIONS: We demonstrated significant temporal changes in retinal morphology in response to nicotine exposure, diabetes and with the combined effects of nicotine and diabetes. These findings may have implications in determining treatment strategies for diabetic patients using products containing nicotine, such as cigarettes, smokeless tobacco, electronic cigarettes or smoking cessation products.


Subject(s)
Cholinergic Agents/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Diabetic Retinopathy/physiopathology , Nicotine/pharmacology , Retina/drug effects , Animals , Cell Survival , Diabetes Mellitus, Experimental/diagnosis , Diabetic Retinopathy/diagnosis , Fluorescein Angiography , Male , Microscopy, Confocal , Multimodal Imaging , Ophthalmoscopy , Rats , Rats, Sprague-Dawley , Retina/pathology , Retinal Neurons/drug effects , Retinal Neurons/pathology , Tomography, Optical Coherence
12.
Biomed Res Int ; 2014: 902842, 2014.
Article in English | MEDLINE | ID: mdl-25162034

ABSTRACT

Accumulating evidence has shown that diabetes accelerates aging and endothelial cell senescence is involved in the pathogenesis of diabetic vascular complications, including diabetic retinopathy. Oxidative stress is recognized as a key factor in the induction of endothelial senescence and diabetic retinopathy. However, specific mechanisms involved in oxidative stress-induced endothelial senescence have not been elucidated. We hypothesized that Sirt6, which is a nuclear, chromatin-bound protein critically involved in many pathophysiologic processes such as aging and inflammation, may have a role in oxidative stress-induced vascular cell senescence. Measurement of Sirt6 expression in human endothelial cells revealed that H2O2 treatment significantly reduced Sirt6 protein. The loss of Sirt6 was associated with an induction of a senescence phenotype in endothelial cells, including decreased cell growth, proliferation and angiogenic ability, and increased expression of senescence-associated ß-galactosidase activity. Additionally, H2O2 treatment reduced eNOS expression, enhanced p21 expression, and dephosphorylated (activated) retinoblastoma (Rb) protein. All of these alternations were attenuated by overexpression of Sirt6, while partial knockdown of Sirt6 expression by siRNA mimicked the effect of H2O2. In conclusion, these results suggest that Sirt6 is a critical regulator of endothelial senescence and oxidative stress-induced downregulation of Sirt6 is likely involved in the pathogenesis of diabetic retinopathy.


Subject(s)
Cellular Senescence/drug effects , Diabetic Retinopathy/genetics , Oxidative Stress/genetics , Sirtuins/biosynthesis , Cellular Senescence/genetics , Diabetic Retinopathy/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/administration & dosage , Nitric Oxide Synthase Type III/biosynthesis , RNA, Small Interfering , Sirtuins/genetics
13.
J Am Heart Assoc ; 3(1): e000476, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24449804

ABSTRACT

BACKGROUND: Development of thoracic aortic aneurysms is the most significant clinical phenotype in patients with Marfan syndrome. An inflammatory response has been described in advanced stages of the disease. Because the hallmark of vascular inflammation is local interleukin-6 (IL-6) secretion, we explored the role of this proinflammatory cytokine in the formation of aortic aneurysms and rupture in hypomorphic fibrillin-deficient mice (mgR/mgR). METHODS AND RESULTS: MgR/mgR mice developed ascending aortic aneurysms with significant dilation of the ascending aorta by 12 weeks (2.7 ± 0.1 and 1.3 ± 0.1 for mgR/mgR versus wild-type mice, respectively; P<0.001). IL-6 signaling was increased in mgR/mgR aortas measured by increases in IL-6 and SOCS3 mRNA transcripts (P<0.05) and in cytokine secretion of IL-6, MCP-1, and GM-CSF (P<0.05). To investigate the role of IL-6 signaling, we generated mgR homozygous mice with IL-6 deficiency (DKO). The extracellular matrix of mgR/mgR mice showed significant disruption of elastin and the presence of dysregulated collagen deposition in the medial-adventitial border by second harmonic generation multiphoton autofluorescence microscopy. DKO mice exhibited less elastin and collagen degeneration than mgR/mgR mice, which was associated with decreased activity of matrix metalloproteinase-9 and had significantly reduced aortic dilation (1.0 ± 0.1 versus 1.6 ± 0.2 mm change from baseline, DKO versus mgR/mgR, P<0.05) that did not affect rupture and survival. CONCLUSION: Activation of IL-6-STAT3 signaling contributes to aneurysmal dilation in mgR/mgR mice through increased MMP-9 activity, aggravating extracellular matrix degradation.


Subject(s)
Aorta/metabolism , Aortic Aneurysm, Thoracic/etiology , Extracellular Matrix/metabolism , Interleukin-6/metabolism , Marfan Syndrome/complications , Microfilament Proteins/metabolism , Animals , Aorta/pathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/prevention & control , Aortic Rupture/etiology , Aortic Rupture/metabolism , Aortic Rupture/pathology , Chemokine CCL2/metabolism , Collagen/metabolism , Dilatation, Pathologic , Disease Models, Animal , Elastin/metabolism , Fibrillin-1 , Fibrillins , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-6/deficiency , Interleukin-6/genetics , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Severity of Illness Index , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors , Up-Regulation
14.
Invest Ophthalmol Vis Sci ; 55(2): 801-13, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24408972

ABSTRACT

PURPOSE: Retinal neovascularization (NV) is a major cause of vision loss in ischemia-induced retinopathy. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor inducible-14 (Fn14), have been implicated in angiogenesis, but their role in retinal diseases is unknown. The goal of this study was to investigate the role of TWEAK/Fn14 pathway in retinal NV. METHODS: Studies were performed in a mouse model of oxygen-induced retinopathy (OIR) and in primary human retinal microvascular endothelial cells (HRMECs). Hyperoxia treatment was initiated on postnatal day (P)14. Immunohistochemistry and quantitative PCR (qPCR) were used to assess retinal vascular changes in relation to expression of Fn14 and TWEAK. RESULTS: Fibroblast growth factor-inducible 14 mRNA was prominently increased from P13 to P17 in OIR retinas, whereas TWEAK level was slightly decreased. These alterations were normalized by hyperoxia treatment and were more striking in isolated retinal vessels. There was a discernible shift in the immunoreactivity of Fn14 and TWEAK from the neuronal layers in the healthy retina to the neovascular tufts in that of OIR. Blockade of TWEAK/Fn14 significantly prevented retinal NV while slightly accelerated revascularization. In contrast, activation of Fn14 positively regulated survival pathways in the B-cell lymphoma-2 (Bcl2) family and robustly enhanced HRMEC survival. Furthermore, gene analysis revealed the regulatory region of Fn14 gene contains several conserved hypoxia inducible factor (HIF)-1α binding sites. Overexpression of HIF-1α prominently induced Fn14 expression in HRMECs. CONCLUSIONS: We found that the TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor inducible-14 (Fn14) pathway is involved in the development of pathologic retinal neovascularization. Hypoxia inducible factor-1α is likely implicated in the upregulation of Fn14.


Subject(s)
Gene Expression Regulation/physiology , Receptors, Tumor Necrosis Factor/genetics , Retinal Neovascularization/genetics , Tumor Necrosis Factors/genetics , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Cytokine TWEAK , Disease Models, Animal , Endothelial Cells/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Regulatory Sequences, Nucleic Acid , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Retinal Vessels/pathology , Signal Transduction/physiology , TWEAK Receptor , Transfection , Tumor Necrosis Factor Inhibitors
15.
Arterioscler Thromb Vasc Biol ; 33(7): 1612-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685554

ABSTRACT

OBJECTIVE: Dysregulated angiotensin II (Ang II) signaling induces local vascular interleukin-6 (IL-6) secretion, producing leukocyte infiltration and life-threatening aortic dissections. Precise mechanisms by which IL-6 signaling induces leukocyte recruitment remain unknown. T-helper 17 lymphocytes (Th17) have been implicated in vascular pathology, but their role in the development of aortic dissections is poorly understood. Here, we tested the relationship of IL-6-signal transducer and activator of transcription-3 signaling with Th17-induced inflammation in the formation of Ang II-induced dissections in C57BL/6 mice. APPROACH AND RESULTS: Ang II infusion induced aortic dissections and CD4(+)-interleukin 17A (IL-17A)-expressing Th17 cell accumulation in C57BL/6 mice. A blunted local Th17 activation, macrophage recruitment, and reduced incidence of aortic dissections were seen in IL-6(-/-) mice. To determine the pathological roles of Th17 lymphocytes, we treated Ang II-infused mice with IL-17A-neutralizing antibody or infused Ang II in genetically deficient IL-17A mice and found decreased aortic chemokine monocytic chemotactic protein-1 production and macrophage recruitment, leading to a reduction in aortic dissections. This effect was independent of blood pressure in IL-17A-neutralizing antibody experiment. Application of a cell-permeable signal transducer and activator of transcription-3 inhibitor to downregulate the IL-6 pathway decreased aortic dilation and Th17 cell recruitment. We also observed increased aortic Th17 infiltration and IL-17 mRNA expression in patients with thoracic aortic dissections. Finally, we found that Ang II-mediated aortic dissections occurred independent of blood pressure changes. CONCLUSIONS: Our results indicate that the IL-6-signal transducer and activator of transcription-3 signaling pathway converges on Th17 recruitment and IL-17A signaling upstream of macrophage recruitment, mediating aortic dissections.


Subject(s)
Angiotensin II , Aorta/immunology , Aortic Aneurysm/immunology , Aortic Dissection/immunology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Th17 Cells/immunology , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/pathology , Aortic Dissection/physiopathology , Aortic Dissection/prevention & control , Animals , Antibodies, Neutralizing/administration & dosage , Aorta/pathology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aortic Aneurysm/physiopathology , Aortic Aneurysm/prevention & control , Blood Pressure , Chemokine CCL2/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammation Mediators/antagonists & inhibitors , Interleukin-17/antagonists & inhibitors , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-6/deficiency , Interleukin-6/genetics , Lymphocyte Activation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Tissue Culture Techniques
16.
Cell Physiol Biochem ; 31(4-5): 659-72, 2013.
Article in English | MEDLINE | ID: mdl-23711492

ABSTRACT

BACKGROUND: Both endoplasmic reticulum (ER) stress, a fundamental cell response associated with stress-initiated unfolded protein response (UPR), and loss of Klotho, an anti-aging hormone linked to NF-κB-induced inflammation, occur in chronic metabolic diseases such as obesity and type 2 diabetes. We investigated if the loss of Klotho is causally linked to increased ER stress. METHODS: We treated human renal epithelial HK-2, alveolar epithelial A549, HEK293, and SH-SH-SY5Y neuroblastoma cells with ER stress-inducing agents, thapsigargin and/or tunicamycin. Effects of overexpression or siRNA-mediated knockdown of Klotho on UPR signaling was investigated by immunoblotting and Real-time PCR. RESULTS: Elevated Klotho levels in HK-2 cells decreased expression of ER stress markers phospho--IRE1, XBP-1s, BiP, CHOP, pJNK, and phospho-p38, all of which were elevated in response to tunicamycin and/or thapsigargin. Similar results were observed using A549 cells for XBP-1s, BiP, and CHOP in response to thapsigargin. Conversely, knockdown of Klotho in HEK 293 cells using siRNA caused further thapsigargin-induced increases in pIRE-1, XBP-1s, and BiP. Klotho overexpression in A549 cells blocked thapsigargin-induced caspase and PARP cleavage and improved cell viability. CONCLUSION: Our data indicate that Klotho has an important role in regulating ER stress and that loss of Klotho is causally linked to ER stress-induced apoptosis.


Subject(s)
Glucuronidase/metabolism , Apoptosis , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Glucuronidase/antagonists & inhibitors , Glucuronidase/genetics , HEK293 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Klotho Proteins , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Thapsigargin/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response , Up-Regulation/drug effects
17.
Exp Diabetes Res ; 2012: 349320, 2012.
Article in English | MEDLINE | ID: mdl-23049540

ABSTRACT

Diabetes and smoking are known risk factors for cataract development. In this study, we evaluated the effect of nicotine on the progression of cataracts in a type 1 diabetic rat model. Diabetes was induced in Sprague-Dawley rats by a single injection of 65 mg/kg streptozotocin. Daily nicotine injections were administered subcutaneously. Forty-five rats were divided into groups of diabetics with and without nicotine treatment and controls with and without nicotine treatment. Progression of lens opacity was monitored using a slit lamp biomicroscope and scores were assigned. To assess whether systemic inflammation played a role in mediating cataractogenesis, we studied serum levels of eotaxin, IL-6, and IL-4. The levels of the measured cytokines increased significantly in nicotine-treated and untreated diabetic animals versus controls and demonstrated a positive trend in the nicotine-treated diabetic rats. Our data suggest the presence of a synergistic relationship between nicotine and diabetes that accelerated cataract formation via inflammatory mediators.


Subject(s)
Cataract/complications , Diabetes Complications/chemically induced , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Eye/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Animals , Cataract/chemically induced , Cataract/immunology , Cataract/physiopathology , Chemokine CCL11/blood , Diabetes Complications/blood , Diabetes Complications/immunology , Diabetes Complications/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Disease Progression , Eye/immunology , Hyperglycemia/etiology , Immunologic Factors/administration & dosage , Immunologic Factors/toxicity , Injections, Subcutaneous , Interleukin-4/blood , Interleukin-6/blood , Male , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Streptozocin
18.
J Cardiovasc Transl Res ; 5(4): 479-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581264

ABSTRACT

Like many diseases, diabetic nephropathy is defined in a histopathological context and studied using reductionist approaches that attempt to ameliorate structural changes. Novel technologies in mass spectrometry-based proteomics have the ability to provide a deeper understanding of the disease beyond classical histopathology, redefine the characteristics of the disease state, and identify novel approaches to reduce renal failure. The goal is to translate these new definitions into improved patient outcomes through diagnostic, prognostic, and therapeutic tools. Here, we review progress made in studying the proteomics of diabetic nephropathy and provide an introduction to the informatics tools used in the analysis of systems biology data, while pointing out statistical issues for consideration. Novel bioinformatics methods may increase biomarker identification, and other tools, including selective reaction monitoring, may hasten clinical validation.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney/metabolism , Proteins/analysis , Proteomics , Systems Biology , Animals , Biomarkers/analysis , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/physiopathology , Diabetic Nephropathies/therapy , Humans , Kidney/physiopathology , Phenotype , Prognosis , Proteomics/methods , Translational Research, Biomedical
19.
Mol Med ; 18: 948-56, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22572938

ABSTRACT

Hyperglycemia and inflammation are hallmarks of burn injury. In this study, we used a rat model of hyperglycemia and burn injury to investigate the effects of hyperglycemia on inflammatory responses in the liver. Hyperglycemia was induced in male Sprague-Dawley rats with streptozotocin (STZ) (35-40 mg/kg), followed by a 60% third-degree scald burn injury. Cytokine levels (by multiplex, in cytosolic liver extracts), hormones (by enzyme-linked immunosorbent assay [ELISA], in serum), nuclear factor (NF)-κB protein deoxyribonucleic acid (DNA) binding (by ELISA, in nuclear liver extracts) and liver functional panel (using VetScan, in serum) were measured at different time points up to 7 d after burn injury. Blood glucose significantly increased after burn injury in both groups with different temporal patterns. Hyperglycemic rats were capable of endogenous insulin secretion, which was enhanced significantly versus controls 12 h after burn injury. DNA binding data of liver nuclear extracts showed a robust and significant activation of the noncanonical NF-κB pathway in the hyperglycemic versus control burn animals, including increased NF-κB-inducing kinase expression (p < 0.05). Liver acute-phase proteins and cytokine expression were increased, whereas secretion of constitutive proteins was decreased after burn injury in hyperglycemic versus control animals (p < 0.05). These results indicate that burn injury to the skin rapidly activated canonical and noncanonical NF-κB pathways in the liver. Robust activation of the NF-κB noncanonical pathway was associated with increased expression of inflammatory markers and acute-phase proteins, and impaired glucose metabolism. Hyperglycemia is detrimental to burn outcome by augmenting inflammation mediated by hepatic noncanonical NF-κB pathway activation.


Subject(s)
Burns/complications , Burns/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Inflammation/complications , Liver/pathology , NF-kappa B/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Blotting, Western , Body Weight , Burns/blood , Cytokines/metabolism , Cytosol/metabolism , DNA/metabolism , Hyperglycemia/blood , Hyperglycemia/pathology , Inflammation/blood , Inflammation/metabolism , Inflammation/pathology , Insulin/metabolism , Insulin Secretion , Liver/metabolism , Male , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , NF-kappaB-Inducing Kinase
20.
Endocrinology ; 152(10): 3622-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21846802

ABSTRACT

Enhanced levels of nuclear factor (NF)-κB-inducing kinase (NIK), an upstream kinase in the NF-κB pathway, have been implicated in the pathogenesis of chronic inflammation in diabetes. We investigated whether increased levels of NIK could induce skeletal muscle insulin resistance. Six obese subjects with metabolic syndrome underwent skeletal muscle biopsies before and six months after gastric bypass surgery to quantitate NIK protein levels. L6 skeletal myotubes, transfected with NIK wild-type or NIK kinase-dead dominant negative plasmids, were treated with insulin alone or with adiponectin and insulin. Effects of NIK overexpression on insulin-stimulated glucose uptake were estimated using tritiated 2-deoxyglucose uptake. NF-κB activation (EMSA), phosphatidylinositol 3 (PI3) kinase activity, and phosphorylation of inhibitor κB kinase ß and serine-threonine kinase (Akt) were measured. After weight loss, skeletal muscle NIK protein was significantly reduced in association with increased plasma adiponectin and enhanced AMP kinase phosphorylation and insulin sensitivity in obese subjects. Enhanced NIK expression in cultured L6 myotubes induced a dose-dependent decrease in insulin-stimulated glucose uptake. The decrease in insulin-stimulated glucose uptake was associated with a significant decrease in PI3 kinase activity and protein kinase B/Akt phosphorylation. Overexpression of NIK kinase-dead dominant negative did not affect insulin-stimulated glucose uptake. Adiponectin treatment inhibited NIK-induced NF-κB activation and restored insulin sensitivity by restoring PI3 kinase activation and subsequent Akt phosphorylation. These results indicate that NIK induces insulin resistance and further indicate that adiponectin exerts its insulin-sensitizing effect by suppressing NIK-induced skeletal muscle inflammation. These observations suggest that NIK could be an important therapeutic target for the treatment of insulin resistance associated with inflammation in obesity and type 2 diabetes.


Subject(s)
Adiponectin/pharmacology , Insulin Resistance , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/physiology , Adult , Cells, Cultured , Glucose/metabolism , Humans , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/analysis , Proto-Oncogene Proteins c-akt/metabolism , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...