Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Neurosci ; 92: 128-136, 2018 10.
Article in English | MEDLINE | ID: mdl-29969654

ABSTRACT

Astrocytes play an indispensable role in maintaining a healthy, functional neural network in the central nervous system (CNS). A primary function of CNS astrocytes is to support the survival and function of neurons. In response to injury, astrocytes take on a reactive phenotype, which alters their molecular functions. Reactive astrocytes have been reported to be both beneficial and harmful to the CNS recovery process subsequent to injury. Understanding the molecular processes and regulatory proteins that determine the extent to which an astrocyte hinders or supports neuronal survival is important within the context of CNS repair. One protein that plays a role in modulating cellular survival is transglutaminase 2 (TG2). Global deletion of TG2 results in beneficial outcomes subsequent to in vivo ischemic brain injury. Ex vivo studies have also implicated TG2 as a negative regulator of astrocyte viability subsequent to injury. In this study we show that knocking down TG2 in astrocytes significantly increases their ability to protect neurons from oxygen glucose deprivation (OGD)/reperfusion injury. To begin to understand how deletion of TG2 in astrocytes improves their ability to protect neurons from injury, we performed transcriptome analysis of wild type and TG2-/- astrocytes. TG2 deletion resulted in alterations in genes involved in extracellular matrix remodeling, cell adhesion and axon growth/guidance. In addition, the majority of genes that showed increases in the TG2-/- astrocytes had predicted cJun/AP-1 binding motifs in their promoters. Furthermore, phospho-cJun levels were robustly elevated in TG2-/- astrocytes, a finding which was consistent with the increase in expression of AP-1 responsive genes. These in vitro data were subsequently extended into an in vivo model to determine whether the absence of astrocytic TG2 improves outcomes after CNS injury. Our results show that, following a spinal cord injury, scar formation is significantly attenuated in mice with astrocyte-specific TG2 deletion compared to mice expressing normal TG2 levels. Taken together, these data indicate that TG2 plays a pivotal role in mediating reactive astrocyte properties following CNS injury. Further, the data suggest that limiting the AP-1 mediated pro-survival injury response may be a contributing factor to that the detrimental effects of astrocytic TG2.


Subject(s)
Astrocytes/metabolism , GTP-Binding Proteins/genetics , Nerve Regeneration , Spinal Cord Injuries/metabolism , Transglutaminases/genetics , Animals , Axon Guidance , Cell Hypoxia , Cells, Cultured , GTP-Binding Proteins/metabolism , Glucose/deficiency , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Spinal Cord Injuries/genetics , Transcription Factor AP-1/metabolism , Transcriptome , Transglutaminases/metabolism
2.
Neuroreport ; 28(18): 1208-1214, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-28957946

ABSTRACT

The multifunctional protein transglutaminase 2 (TG2) has been widely implicated as a modulator of cellular viability. Specifically, TG2 expression is beneficial to neuronal survival following an ischemic injury, whereas the opposite is true in astrocytes. Furthermore, its role in mediating cell death and survival processes has been suggested to be dependent on its subcellular localization. Therefore, the aim of this study was to examine the subcellular localization patterns of neuronal and astrocytic TG2 in ischemia-relevant conditions. We found that nuclear levels of TG2 were significantly increased in neurons, but reduced in astrocytes, in response to hypoxia. In addition, there were no changes in extracellular TG2 in astrocytes exposed to hypoxia. Thus, these findings demonstrate a difference in the subcellular localization pattern of TG2 in neurons and astrocytes in ischemia-relevant conditions and provide further avenues for investigation into the role of TG2 in mediating cellular viability.


Subject(s)
Astrocytes/ultrastructure , GTP-Binding Proteins/metabolism , Hypoxia/pathology , Neurons/ultrastructure , Subcellular Fractions/enzymology , Transglutaminases/metabolism , Animals , Animals, Newborn , Biotinylation , Cell Nucleus/enzymology , Cells, Cultured , Cerebral Cortex , Culture Media, Serum-Free , Cytosol/enzymology , Female , Hypoxia/physiopathology , Mice , Pregnancy , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Sprague-Dawley
3.
Brain Res ; 1668: 1-11, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28522262

ABSTRACT

Transglutaminase 2 (TG2) is a multifunctional protein that can contribute to cell death and cell survival processes in a variety of disease contexts. Within the brain, TG2 has been shown to promote cell death in ischemic injury when expressed in astrocytes (Colak and Johnson, 2012). However, the specific functions and characteristics of astrocytic TG2 that mediate this effect are largely unknown. Therefore, the goal of this study was to investigate the role of astrocytic TG2 in mediating cellular viability processes in the context of ischemic injury, with a specific focus on its contributions to intracellular signaling cascades. We show that, in response to oxygen/glucose deprivation (OGD), acute lentiviral-mediated knockdown of TG2, as well as inhibition with an irreversible TG2 inhibitor, enhances cell survival. We also show that TG2 depletion increases nuclear factor-κB (NF-κB) signaling, whereas inhibition reduces NF-κB activity. Despite its clear contribution to NF-κB signaling, however, TG2 modulation of NF-κB signaling is not likely to be a major contributor to its ability to mediate astrocytic viability in this context. Overall, the results of this study provide insight into the role of TG2 in astrocytes and suggest possible avenues for future study of the relationship between astrocytic TG2 and ischemic injury.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , GTP-Binding Proteins/metabolism , Ischemia/metabolism , NF-kappa B/metabolism , Signal Transduction , Transglutaminases/metabolism , Animals , Brain/metabolism , Cell Death/genetics , Cell Survival/genetics , Cells, Cultured , Ischemia/genetics , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Protein Glutamine gamma Glutamyltransferase 2 , Signal Transduction/physiology
4.
Kidney Int ; 89(6): 1281-92, 2016 06.
Article in English | MEDLINE | ID: mdl-27165830

ABSTRACT

Endostatin (EST), an antiangiogenic factor, is enriched in aging kidneys. EST is also an interactive partner of transglutaminase 2 (TG2), an enzyme that cross-links extracellular matrix proteins. Here we tested whether EST and TG2 play a role in the fibrosis of aging. In wild-type mice, aging kidneys exhibited a 2- to 4-fold increase in TG2 paralleled by increased cross-linked extracellular matrix proteins and fibrosis. Mice transgenic to express EST showed renal fibrosis at a young age. One-month delivery of EST via minipumps to young mice showed increased renal fibrosis that became more robust when superimposed on folic acid-induced nephropathy. Upregulated TG2 and impaired renal function were apparent with EST delivery combined with folic acid-induced nephropathy. Subcapsular injection of TG2 and/or EST into kidneys of young mice not only induced interstitial fibrosis, but also increased the proportion of senescent cells. Thus, kidney fibrosis in aging may represent a natural outcome of upregulated EST and TG2, but more likely it appears to be a result of cumulative stresses occurring on the background of synergistically acting geronic (aging) proteins, EST and TG2.


Subject(s)
Aging/metabolism , Collagen Type XVIII/metabolism , Endostatins/metabolism , GTP-Binding Proteins/metabolism , Kidney Diseases/pathology , Kidney/pathology , Transglutaminases/metabolism , Animals , Cells, Cultured , Cellular Senescence/drug effects , Collagen Type XVIII/genetics , Collagen Type XVIII/pharmacology , Endostatins/genetics , Endostatins/pharmacology , Endothelial Cells , Extracellular Matrix Proteins , Fibrosis , Folic Acid/toxicity , GTP-Binding Proteins/genetics , GTP-Binding Proteins/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/genetics , Transglutaminases/pharmacology , Up-Regulation
5.
Biochim Biophys Acta ; 1833(1): 1-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23085038

ABSTRACT

Transglutaminase 2 (TG2) is a widely expressed and multifunctional protein that modulates cell death/survival processes. We have previously shown that TG2 binds to hypoxia inducible factor 1ß (HIF1ß) and decreases the upregulation of HIF responsive genes; however, the relationship between these observations was not investigated. In this study, we investigated whether endogenous TG2 is sufficient to suppress HIF activity and whether the interaction between TG2 and HIF1ß is required for this suppression. shRNA-mediated silencing of TG2 significantly enhanced HIF activation in response to hypoxia. In addition, nuclear localization of TG2 is required for its suppressive effect on HIF activity, with TG2 being recruited to HIF responsive promoters in hypoxic conditions. These observations suggest that TG2 directly regulates hypoxic transcriptional machinery; however, its interaction with HIF1ß was not required for this regulation. We also examined whether TG2's effect on cell death/survival processes in ischemia is due to its effects on HIF signaling. Our results indicate that TG2 mediated HIF suppression can be separated from TG2's effect on cell survival in hypoxic/hypoglycemic conditions. Lastly, here we show that nuclear TG2 in the closed conformation and non-nuclear TG2 in the open conformation have opposing effects on hypoxic/hypoglycemic cell death, which could explain previous controversial results. Overall, our results further clarify the role of TG2 in mediating the cellular response to ischemia and suggest that manipulating the conformation of TG2 might be of pharmacological interest as a therapeutic strategy for the treatment of ischemia-related pathologies.


Subject(s)
GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transglutaminases/chemistry , Transglutaminases/metabolism , Transglutaminases/physiology , Animals , Cell Death/genetics , Cell Death/physiology , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Nucleus/metabolism , Cells, Cultured , GTP-Binding Proteins/genetics , HEK293 Cells , Humans , Hypoglycemia/genetics , Hypoglycemia/metabolism , Hypoglycemia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Neurons/metabolism , Protein Conformation , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport/physiology , Rats , Signal Transduction/genetics , Signal Transduction/physiology , Structure-Activity Relationship , Transglutaminases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...