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










Publication year range
1.
Exp Neurol ; 339: 113646, 2021 05.
Article in English | MEDLINE | ID: mdl-33600817

ABSTRACT

Spliced X-box binding protein-1 (XBP1s) together with the hexosamine biosynthetic pathway (HBP) and O-GlcNAcylation forms the XBP1s/HBP/O-GlcNAc axis. Our previous studies have provided evidence that activation of this axis is neuroprotective after ischemic stroke and critically, ischemia-induced O-GlcNAcylation is impaired in the aged brain. However, the XBP1s' neuroprotective role and its link to O-GlcNAcylation in stroke, as well as the therapeutic potential of targeting this axis in stroke, have not been well established. Moreover, the mechanisms underlying this age-related impairment of O-GlcNAcylation induction after brain ischemia remain completely unknown. In this study, using transient ischemic stroke models, we first demonstrated that neuron-specific overexpression of Xbp1s improved outcome, and pharmacologically boosting O-GlcNAcylation with thiamet-G reversed worse outcome observed in neuron-specific Xbp1 knockout mice. We further showed that thiamet-G treatment improved long-term functional recovery in both young and aged animals after transient ischemic stroke. Mechanistically, using an analytic approach developed here, we discovered that availability of UDP-GlcNAc was compromised in the aged brain, which may constitute a novel mechanism responsible for the impaired O-GlcNAcylation activation in the aged brain after ischemia. Finally, based on this new mechanistic finding, we evaluated and confirmed the therapeutic effects of glucosamine treatment in young and aged animals using both transient and permanent stroke models. Our data together support that increasing O-GlcNAcylation is a promising strategy in stroke therapy.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Brain/metabolism , Ischemic Stroke/metabolism , Ischemic Stroke/prevention & control , Neuroprotection/physiology , Age Factors , Animals , Brain/drug effects , Glucosamine/pharmacology , Glucosamine/therapeutic use , Glycosylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats , Rats, Inbred F344 , X-Box Binding Protein 1/deficiency , X-Box Binding Protein 1/genetics
2.
FASEB J ; 34(11): 14750-14767, 2020 11.
Article in English | MEDLINE | ID: mdl-32910521

ABSTRACT

Small ubiquitin-like modifier (SUMO1-3) conjugation (SUMOylation), a posttranslational modification, modulates almost all major cellular processes. Mounting evidence indicates that SUMOylation plays a crucial role in maintaining and regulating neural function, and importantly its dysfunction is implicated in cognitive impairment in humans. We have previously shown that simultaneously silencing SUMO1-3 expression in neurons negatively affects cognitive function. However, the roles of the individual SUMOs in modulating cognition and the mechanisms that link SUMOylation to cognitive processes remain unknown. To address these questions, in this study, we have focused on SUMO2 and generated a new conditional Sumo2 knockout mouse line. We found that conditional deletion of Sumo2 predominantly in forebrain neurons resulted in marked impairments in various cognitive tests, including episodic and fear memory. Our data further suggest that these abnormalities are attributable neither to constitutive changes in gene expression nor to alterations in neuronal morphology, but they involve impairment in dynamic SUMOylation processes associated with synaptic plasticity. Finally, we provide evidence that dysfunction on hippocampal-based cognitive tasks was associated with a significant deficit in the maintenance of hippocampal long-term potentiation in Sumo2 knockout mice. Collectively, these data demonstrate that protein conjugation by SUMO2 is critically involved in cognitive processes.


Subject(s)
Memory , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Cognition , Female , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Prosencephalon/metabolism , Prosencephalon/physiology , Small Ubiquitin-Related Modifier Proteins/genetics
3.
Stroke ; 51(5): 1570-1577, 2020 05.
Article in English | MEDLINE | ID: mdl-32212900

ABSTRACT

Background and Purpose- Ischemic stroke impairs endoplasmic reticulum (ER) function, causes ER stress, and activates the unfolded protein response. The unfolded protein response consists of 3 branches controlled by ER stress sensor proteins, which include PERK (protein kinase RNA-like ER kinase). Activated PERK phosphorylates eIF2α (eukaryotic initiation factor 2 alpha), resulting in inhibition of global protein synthesis. Here, we aimed to clarify the role of the PERK unfolded protein response branch in stroke. Methods- Neuron-specific and tamoxifen-inducible PERK conditional knockout (cKO) mice were generated by cross-breeding Camk2a-CreERT2 with Perkf/f mice. Transient middle cerebral artery occlusion was used to induce stroke. Short- and long-term stroke outcomes were evaluated. Protein synthesis in the brain was assessed using a surface-sensing-of-translation approach. Results- After tamoxifen-induced deletion of Perk in forebrain neurons was confirmed in PERK-cKO mice, PERK-cKO and control mice were subjected to transient middle cerebral artery occlusion and 3 days or 3 weeks recovery. PERK-cKO mice had larger infarcts and worse neurological outcomes compared with control mice, suggesting that PERK-induced eIF2α phosphorylation and subsequent suppression of translation protects neurons from ischemic stress. Indeed, better stroke outcomes were observed in PERK-cKO mice that received postischemic treatment with salubrinal, which can restore the ischemia-induced increase in phosphorylated eIF2α in these mice. Finally, our data showed that post-treatment with salubrinal improved functional recovery after stroke. Conclusions- Here, we presented the first evidence that postischemic suppression of translation induced by PERK activation promotes recovery of neurological function after stroke. This confirms and further extends our previous observations that recovery of ER function impaired by ischemic stress critically contributes to stroke outcome. Therefore, future research should include strategies to improve stroke outcome by targeting unfolded protein response branches to restore protein homeostasis in neurons.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/metabolism , Neuroprotection/genetics , Unfolded Protein Response/genetics , eIF-2 Kinase/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cinnamates/pharmacology , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/drug effects , Infarction, Middle Cerebral Artery/physiopathology , Mice , Mice, Knockout , Phosphorylation , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Stroke/metabolism , Stroke/physiopathology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Unfolded Protein Response/drug effects
4.
Dev Dyn ; 249(2): 237-244, 2020 02.
Article in English | MEDLINE | ID: mdl-31625212

ABSTRACT

Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is essential for the development of healthy placenta. The loss of SENP2 causes severe placental deficiencies and leads to embryonic death that is associated with heart and brain deformities. However, tissue-specific disruption of SENP2 demonstrates its dispensable role in embryogenesis and the embryonic defects are secondary to placental insufficiency. SENP2 regulates SUMO1 modification of Mdm2, which controls p53 activities critical for trophoblast cell proliferation and differentiation. Here we use genetic analyses to examine the involvement of SUMO2 and SUMO3 for SENP2-mediated placentation. The results indicate that hyper-SUMOylation caused by SENP2 deficiency can be compensated by reducing the level of SUMO modifiers. The placental deficiencies caused by the loss of SENP2 can be alleviated by the inactivation of gene encoding SUMO2 or SUMO3. Our findings demonstrate that SENP2 genetically interacts with SUMO2 and SUMO3 pivotal for the development of three major trophoblast layers. The alleviation of placental defects in the SENP2 knockouts further leads to the proper formation of the heart structures, including atrioventricular cushion and myocardium. SUMO2 and SUMO3 modifications regulate placentation and organogenesis mediated by SENP2.


Subject(s)
Cysteine Endopeptidases/metabolism , Embryo, Mammalian/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Animals , Cysteine Endopeptidases/genetics , Female , Mice , Mice, Knockout , Placental Insufficiency/genetics , Placental Insufficiency/metabolism , Pregnancy , Small Ubiquitin-Related Modifier Proteins/genetics , Trophoblasts/metabolism , Ubiquitins/genetics
5.
J Am Heart Assoc ; 7(17): e009634, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30371162

ABSTRACT

Background The mechanisms underlying worse outcome at advanced age after cardiac arrest ( CA ) and resuscitation are not well understood. Because protein homeostasis (proteostasis) is essential for cellular and organismal health, but is impaired after CA , we investigated the effects of age on proteostasis-related prosurvival pathways activated after CA . Methods and Results Young (2-3 months old) and aged (21-22 months old) male C57Bl/6 mice were subjected to CA and cardiopulmonary resuscitation ( CPR ). Functional outcome and organ damage were evaluated by assessing neurologic deficits, histological features, and creatinine level. CA / CPR -related changes in small ubiquitin-like modifier conjugation, ubiquitination, and the unfolded protein response were analyzed by measuring mRNA and protein levels in the brain, kidney, and spinal cord. Thiamet-G was used to increase O-linked ß-N-acetylglucosamine modification. After CA / CPR , aged mice had trended lower survival rates, more severe tissue damage in the brain and kidney, and poorer recovery of neurologic function compared with young mice. Furthermore, small ubiquitin-like modifier conjugation, ubiquitination, unfolded protein response, and O-linked ß-N-acetylglucosamine modification were activated after CA / CPR in young mice, but their activation was impaired in aged mice. Finally, pharmacologically increasing O-linked ß-N-acetylglucosamine modification after CA improved outcome. Conclusions Results suggest that impaired activation of prosurvival pathways contributes to worse outcome after CA / CPR in aged mice because restoration of proteostasis is critical to the survival of cells stressed by ischemia. Therefore, a pharmacologic intervention that targets aging-related impairment of proteostasis-related pathways after CA / CPR may represent a promising therapeutic strategy.


Subject(s)
Aging/metabolism , Brain/metabolism , Heart Arrest/metabolism , Kidney/metabolism , Spinal Cord/metabolism , Unfolded Protein Response , Acetylglucosamine/metabolism , Animals , Brain/pathology , Cardiopulmonary Resuscitation , Heart Arrest/physiopathology , Heart Arrest/therapy , Kidney/pathology , Mice , Proteostasis , Recovery of Function , Small Ubiquitin-Related Modifier Proteins/metabolism , Spinal Cord/pathology , Ubiquitination
6.
Lab Invest ; 98(6): 799-813, 2018 06.
Article in English | MEDLINE | ID: mdl-29472640

ABSTRACT

The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.


Subject(s)
Intestinal Mucosa/blood supply , Reperfusion Injury/prevention & control , SUMO-1 Protein/physiology , Ubiquitin-Conjugating Enzymes/physiology , Animals , Chemokines/analysis , Intestinal Mucosa/chemistry , Laser Capture Microdissection , Mice , Mice, Inbred C57BL , Mice, Knockout , SUMO-1 Protein/deficiency , Small Ubiquitin-Related Modifier Proteins/analysis , Small Ubiquitin-Related Modifier Proteins/physiology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/analysis , Ubiquitins/physiology
7.
Aging Dis ; 9(1): 31-39, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29392079

ABSTRACT

Experimental cardiac arrest (CA) in aging research is infrequently studied in part due to the limitation of animal models. We aimed to develop an easily performed mouse CA model to meet this need. A standard mouse KCl-induced CA model using chest compressions and intravenous epinephrine for resuscitation was modified by blood withdrawal prior to CA onset, so as to decrease the requisite KCl dose to induce CA by decreasing the circulating blood volume. The modification was then compared to the standard model in young adult mice subjected to 8 min CA. 22-month old mice were then subjected to 8 min CA, resuscitated, and compared to young adult mice. Post-CA functional recovery was evaluated by measuring spontaneous locomotor activity pre-injury, and on post-CA days 1, 2, and 3. Neurological score and brain histology were examined on day 3. Brain elF2α phosphorylation levels were measured at 1 h to verify tissue stress. Compared to the standard model, the modification decreased cardiopulmonary resuscitation duration and increased 3-day survival in young mice. For aged mice, survival was 100 % at 24 h and 54% at 72 h. Neurological deficit was present 3 days post-CA, although more severe versus young mice. Mild neuronal necrosis was present in the cortex and hippocampus. The modified model markedly induced elF2α phosphorylation in both age groups. This modified procedure makes the CA model feasible in aged mice and provides a practical platform for understanding injury mechanisms and developing therapeutics for elderly patients.

8.
J Cereb Blood Flow Metab ; 38(1): 5-16, 2018 01.
Article in English | MEDLINE | ID: mdl-29148315

ABSTRACT

Post-translational protein modification by small ubiquitin-like modifier (SUMO) regulates a myriad of homeostatic and stress responses. The SUMOylation pathway has been extensively studied in brain ischemia. Convincing evidence is now at hand to support the notion that a major increase in levels of SUMOylated proteins is capable of inducing tolerance to ischemic stress. Therefore, the SUMOylation pathway has emerged as a promising therapeutic target for neuroprotection in the face of brain ischemia. Despite this, it is prudent to acknowledge that there are many key questions still to be addressed in brain ischemia related to SUMOylation. Accordingly, herein, we provide a critical review of literature within the field to summarize current knowledge and in so doing highlight pertinent translational implications of the SUMOylation pathway in brain ischemia.


Subject(s)
Brain Ischemia/metabolism , Neuroprotection/physiology , Sumoylation/physiology , Humans
9.
J Cereb Blood Flow Metab ; 37(10): 3318-3324, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28752781

ABSTRACT

Neuroprotection strategies to improve stroke outcome have been successful in the laboratory but not in clinical stroke trials, and thus have come under scrutiny by the medical community. Experimental stroke investigators are therefore under increased pressure to resolve this problem. Acute ischemic stroke represents a severe form of metabolic stress that activates many pathological processes and thereby impairs cellular functions. Traditionally, neuroprotection strategies were designed to improve stroke outcome by interfering with pathological processes triggered by ischemia. However, stroke outcome is also dependent on the brain's capacity to restore cellular functions impaired by ischemia, and this capacity declines with age. It is, therefore, conceivable that this age-dependent decline in the brain's self-healing capacity contributes to the disparity between the success of neuroprotective strategies in young animals, and limited success in elderly stroke patients. Here, prosurvival pathways that restore protein homeostasis impaired by ischemic stress should be considered, because their capacity decreases with increasing age, and maintenance of proteome fidelity is pivotal for cell survival. Boosting such prosurvival pathways pharmacologically to restore protein homeostasis and, thereby, cellular functions impaired by ischemic stress is expected to counterbalance the compromised self-healing capacity of aged brains and thereby help to improve stroke outcome.


Subject(s)
Homeostasis , Neuroprotective Agents/pharmacology , Proteome/physiology , Stroke/physiopathology , Age Factors , Animals , Brain Ischemia/physiopathology , Humans , Neuroprotective Agents/therapeutic use
10.
Stroke ; 48(6): 1646-1654, 2017 06.
Article in English | MEDLINE | ID: mdl-28487326

ABSTRACT

BACKGROUND AND PURPOSE: Impaired protein homeostasis induced by endoplasmic reticulum dysfunction is a key feature of a variety of age-related brain diseases including stroke. To restore endoplasmic reticulum function impaired by stress, the unfolded protein response is activated. A key unfolded protein response prosurvival pathway is controlled by the endoplasmic reticulum stress sensor (inositol-requiring enzyme-1), XBP1 (downstream X-box-binding protein-1), and O-GlcNAc (O-linked ß-N-acetylglucosamine) modification of proteins (O-GlcNAcylation). Stroke impairs endoplasmic reticulum function, which activates unfolded protein response. The rationale of this study was to explore the potentials of the IRE1/XBP1/O-GlcNAc axis as a target for neuroprotection in ischemic stroke. METHODS: Mice with Xbp1 loss and gain of function in neurons were generated. Stroke was induced by transient or permanent occlusion of the middle cerebral artery in young and aged mice. Thiamet-G was used to increase O-GlcNAcylation. RESULTS: Deletion of Xbp1 worsened outcome after transient and permanent middle cerebral artery occlusion. After stroke, O-GlcNAcylation was activated in neurons of the stroke penumbra in young mice, which was largely Xbp1 dependent. This activation of O-GlcNAcylation was impaired in aged mice. Pharmacological increase of O-GlcNAcylation before or after stroke improved outcome in both young and aged mice. CONCLUSIONS: Our study indicates a critical role for the IRE1/XBP1 unfolded protein response branch in stroke outcome. O-GlcNAcylation is a prosurvival pathway that is activated in the stroke penumbra in young mice but impaired in aged mice. Boosting prosurvival pathways to counterbalance the age-related decline in the brain's self-healing capacity could be a promising strategy to improve ischemic stroke outcome in aged brains.


Subject(s)
Acetylglucosamine/metabolism , Brain Ischemia/metabolism , Membrane Proteins/metabolism , Neuroprotection/physiology , Protein Serine-Threonine Kinases/metabolism , Pyrans/pharmacology , Stroke/metabolism , Thiazoles/pharmacology , Unfolded Protein Response/physiology , X-Box Binding Protein 1/metabolism , Age Factors , Animals , Disease Models, Animal , Infarction, Middle Cerebral Artery , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Folding
11.
Neuroscience ; 343: 190-212, 2017 02 20.
Article in English | MEDLINE | ID: mdl-27919694

ABSTRACT

Small ubiquitin-like modifier (SUMO) conjugation (SUMOylation) plays key roles in neurologic function in health and disease. Neuronal SUMOylation is essential for emotionality and cognition, and this pathway is dramatically activated in post-ischemic neurons, a neuroprotective response to ischemia. It is also known from cell culture studies that SUMOylation modulates gene expression. However, it remains unknown how SUMOylation regulates neuronal gene expression in vivo, in the physiologic state and after ischemia, and modulates post-ischemic recovery of neurologic function. To address these important questions, we used a SUMO1-3 knockdown (SUMO-KD) mouse in which a Thy-1 promoter drives expression of 3 distinct microRNAs against SUMO1-3 to silence SUMO expression specifically in neurons. Wild-type and SUMO-KD mice were subjected to transient forebrain ischemia. Microarray analysis was performed in hippocampal CA1 samples, and neurologic function was evaluated. SUMOylation had opposite effects on neuronal gene expression before and after ischemia. In the physiological state, most genes regulated by SUMOylation were up-regulated in SUMO-KD compared to wild-type mice. Brain ischemia/reperfusion significantly modulated the expression levels of more than 400 genes in wild-type mice, with a majority of those genes upregulated. The extent of this post-ischemic transcriptome change was suppressed in SUMO-KD mice. Moreover, SUMO-KD mice exhibited significantly worse functional outcome. This suggests that suppression of global gene expression response in post-ischemic brain due to SUMO knockdown has a negative effect on post-ischemic neurologic function. Together, our data provide a basis for future studies to mechanistically link SUMOylation to neurologic function in health and disease.


Subject(s)
Brain Ischemia/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Recovery of Function/physiology , Small Ubiquitin-Related Modifier Proteins/deficiency , Animals , Blotting, Western , Brain Ischemia/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation/physiology , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Microscopy, Confocal , Motor Activity/physiology , Neurons/pathology , Prosencephalon/pathology , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Small Ubiquitin-Related Modifier Proteins/genetics
12.
J Cereb Blood Flow Metab ; 37(3): 1069-1079, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27217380

ABSTRACT

Impaired function of the endoplasmic reticulum (ER stress) is a hallmark of many human diseases including stroke. To restore ER function in stressed cells, the unfolded protein response (UPR) is induced, which activates 3 ER stress sensor proteins including activating transcription factor 6 (ATF6). ATF6 is then cleaved by proteases to form the short-form ATF6 (sATF6), a transcription factor. To determine the extent to which activation of the ATF6 UPR branch defines the fate and function of neurons after stroke, we generated a conditional and tamoxifen-inducible sATF6 knock-in mouse. To express sATF6 in forebrain neurons, we crossed our sATF6 knock-in mouse line with Emx1-Cre mice to generate ATF6-KI mice. After the ATF6 branch was activated in ATF6-KI mice with tamoxifen, mice were subjected to transient middle cerebral artery occlusion. Forced activation of the ATF6 UPR branch reduced infarct volume and improved functional outcome at 24 h after stroke. Increased autophagic activity at early reperfusion time after stroke may contribute to the ATF6-mediated neuroprotection. We concluded that the ATF6 UPR branch is crucial to ischemic stroke outcome. Therefore, boosting UPR pro-survival pathways may be a promising therapeutic strategy for stroke.


Subject(s)
Activating Transcription Factor 6/metabolism , Neurons/metabolism , Neuroprotection/drug effects , Stroke/drug therapy , Unfolded Protein Response , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/pharmacology , Animals , Autophagy , Brain Infarction , Gene Knock-In Techniques , Infarction, Middle Cerebral Artery , Mice , Recovery of Function
13.
J Cereb Blood Flow Metab ; 36(12): 2044-2050, 2016 12.
Article in English | MEDLINE | ID: mdl-27733676

ABSTRACT

Folding and processing newly synthesized proteins are vital functions of the endoplasmic reticulum that are sensitive to a variety of stress conditions. The unfolded protein response is activated to restore endoplasmic reticulum function impaired by stress. While we know that brain ischemia impairs endoplasmic reticulum function, the role of unfolded protein response activation in post-ischemic recovery of neurologic function is only beginning to emerge. Here, we summarize what is known about endoplasmic reticulum stress and unfolded protein response in brain ischemia and discuss recent findings from myocardial ischemia studies that could help to advance research on endoplasmic reticulum stress and unfolded protein response in brain ischemia.


Subject(s)
Brain Ischemia/pathology , Unfolded Protein Response/physiology , Endoplasmic Reticulum Stress , Humans , Recovery of Function
14.
J Cereb Blood Flow Metab ; 36(2): 393-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26661187

ABSTRACT

To evaluate the effect of age on the response of brains to an ischemic challenge, we subjected young and aged mice to transient forebrain ischemia, and analyzed the heat shock response and unfolded protein response, ubiquitin conjugation and SUMO conjugation, and O-linked ß-N-acetylglucosamine modification of proteins (O-GlcNAcylation). The most prominent age-related difference was an inability of aged mice to activate O-GlcNAcylation. Considering many reports on the protective role of O-GlcNAcylation in various stress conditions including myocardial ischemia, this pathway could be a promising target for therapeutic intervention to improve functional recovery of aged patients following brain ischemia.


Subject(s)
Acetylglucosamine/metabolism , Aging/pathology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/psychology , Brain/pathology , Protein Processing, Post-Translational/genetics , Stroke/pathology , Stroke/psychology , Animals , Brain Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Myocardial Ischemia/genetics , Myocardial Ischemia/physiopathology , Psychomotor Performance , Recovery of Function , SUMO-1 Protein/metabolism , Stress, Physiological/genetics , Ubiquitination , Unfolded Protein Response/genetics
15.
Proteomics ; 15(5-6): 1181-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25236368

ABSTRACT

Small ubiquitin-like modifier (SUMO1-3) conjugation is a posttranslational protein modification whereby SUMOs are conjugated to lysine residues of target proteins. SUMO conjugation can alter the activity, stability, and function of target proteins, and thereby modulate almost all major cellular pathways. Many diseases are associated with SUMO conjugation, including heart failure, arthritis, cancer, degenerative diseases, and brain ischemia/stroke. It is, therefore, of major interest to characterize the SUMO-modified proteome regulated by these disorders. SUMO proteomics analysis is hampered by low levels of SUMOylated proteins. Several strategies have, therefore, been developed to enrich SUMOylated proteins from cell/tissue extracts. These include proteomics analysis on cells expressing epitope-tagged SUMO isoforms, use of monoclonal SUMO antibodies for immunoprecipitation and epitope-specific peptides for elution, and affinity purification with peptides containing SUMO interaction motifs to specifically enrich polySUMOylated proteins. Recently, two mouse models were generated and characterized that express tagged SUMO isoforms, and allow purification of SUMOylated proteins from complex organ extracts. Ultimately, these new analytical tools will help to decipher the SUMO-modified proteome regulated by various human diseases, and thereby, identify new targets for preventive and therapeutic purposes.


Subject(s)
Heart Diseases/metabolism , Neoplasms/metabolism , Proteome , Proteomics/methods , SUMO-1 Protein , Sumoylation/physiology , Animals , Biomarkers , Humans , Mice , Neurodegenerative Diseases/metabolism , Proteome/analysis , Proteome/metabolism , Proteome/physiology , SUMO-1 Protein/analysis , SUMO-1 Protein/metabolism , SUMO-1 Protein/physiology , Stroke/metabolism
16.
J Proteome Res ; 13(9): 3905-18, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25072996

ABSTRACT

SUMOylation is an essential posttranslational modification and regulates many cellular processes. Dysregulation of SUMOylation plays a critical role in metastasis, yet how its perturbation affects this lethal process of cancer is not well understood. We found that SUMO-2/3 modification is greatly up-regulated in metastatic breast cancer cells compared with nonmetastatic control cells. To identify proteins differentially modified by SUMO-2/3 between metastatic and nonmetastatic cells, we established a method in which endogenous SUMO-2/3 conjugates are labeled by stable isotope labeling by amino acids in cell culture (SILAC), immunopurified by SUMO-2/3 monoclonal antibodies and epitope-peptide elution, and analyzed by quantitative mass spectrometry. We identified 66 putative SUMO-2/3-conjugated proteins, of which 15 proteins show a significant increase/decrease in SUMO-2/3 modification in metastatic cells. Targets with altered SUMOylation are involved in cell cycle, migration, inflammation, glycolysis, gene expression, and SUMO/ubiquitin pathways, suggesting that perturbations of SUMO-2/3 modification might contribute to metastasis by affecting these processes. Consistent with this, up-regulation of PML SUMO-2/3 modification corresponds to an increased number of PML nuclear bodies (PML-NBs) in metastatic cells, whereas up-regulation of global SUMO-2/3 modification promotes 3D cell migration. Our findings provide a foundation for further investigating the effects of SUMOylation on breast cancer progression and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proteomics/methods , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Breast Neoplasms/chemistry , Cell Line, Tumor , Disease Progression , Drosophila , Female , Humans , Mass Spectrometry , Mice , Neoplastic Processes , Protein Processing, Post-Translational , Sequence Alignment , Small Ubiquitin-Related Modifier Proteins/analysis
17.
EMBO Rep ; 15(8): 878-85, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891386

ABSTRACT

Small ubiquitin-like modifier (SUMO1-3) conjugation plays a critical role in embryogenesis. Embryos deficient in the SUMO-conjugating enzyme Ubc9 die at the early postimplantation stage. Sumo1(-/-) mice are viable, as SUMO2/3 can compensate for most SUMO1 functions. To uncover the role of SUMO2/3 in embryogenesis, we generated Sumo2- and Sumo3-null mutant mice. Here, we report that Sumo3(-/-) mice were viable, while Sumo2(-/-) embryos exhibited severe developmental delay and died at approximately embryonic day 10.5 (E10.5). We also provide evidence that SUMO2 is the predominantly expressed SUMO isoform. Furthermore, although Sumo2(+/-) and Sumo2(+/-);Sumo3(+/-) mice lacked any overt phenotype, only 2 Sumo2(+/-);Sumo3(-/-) mice were found at birth in 35 litters after crossing Sumo2(+/-);Sumo3(+/-) with Sumo3(-/-) mice, and these rare mice were considerably smaller than littermates of the other genotypes. Thus, our findings suggest that expression levels and not functional differences between SUMO2 and SUMO3 are critical for normal embryogenesis.


Subject(s)
Embryonic Development , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitins/genetics , Animals , Female , Gene Expression , Genes, Essential , Mice, Inbred C57BL , Mice, Knockout , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism
18.
J Psychiatry Neurosci ; 39(4): 259-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24690371

ABSTRACT

BACKGROUND: Growing evidence suggests that small ubiquitin-like modifier (SUMO) conjugation plays a key role in brain plasticity by modulating activity-dependent synaptic transmission. However, these observations are based largely on cell culture experiments. We hypothesized that episodic and fear memories would be affected by silencing SUMO1-3 expression. METHODS: To investigate the role of SUMO conjugation in neuronal functioning in vivo, we generated a novel Sumo transgenic mouse model in which a Thy1 promoter drives expression of 3 distinct microRNAs to silence Sumo1-3 expression, specifically in neurons. Wild-type and Sumo1-3 knockdown mice were subjected to a battery of behavioural tests to elucidate whether Sumoylation is involved in episodic and emotional memory. RESULTS: Expression of Sumo1-3 microRNAs and the corresponding silencing of Sumo expression were particularly pronounced in hippocampal, amygdala and layer V cerebral cortex neurons. The Sumo knockdown mice displayed anxiety-like responses and were impaired in episodic memory processes, contextual and cued fear conditioning and fear-potentiated startle. LIMITATIONS: Since expression of Sumo1-3 was silenced in this mouse model, we need to verify in future studies which of the SUMO paralogues play the pivotal role in episodic and emotional memory. CONCLUSION: Our results indicate that a functional SUMO conjugation pathway is essential for emotionality and cognition. This novel Sumo knockdown mouse model and the technology used in generating this mutant may help to reveal novel mechanisms that underlie a variety of neuropsychiatric conditions associated with anxiety and impairment of episodic and emotional memory.


Subject(s)
Fear/physiology , Memory/physiology , Neurons/physiology , Animals , Anxiety/physiopathology , Brain/physiopathology , Conditioning, Psychological/physiology , Cues , Emotions/physiology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Gene Silencing , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Neuropsychological Tests , Reflex, Startle/physiology
19.
Stroke ; 45(4): 1115-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569813

ABSTRACT

BACKGROUND AND PURPOSE: Small ubiquitin-like modifier (SUMO) conjugation is a post-translational modification associated with many human diseases. Characterization of the SUMO-modified proteome is pivotal to define the mechanistic link between SUMO conjugation and such diseases. This is particularly evident for SUMO2/3 conjugation, which is massively activated after brain ischemia/stroke, and is believed to be a protective response. The purpose of this study was to perform a comprehensive analysis of the SUMO3-modified proteome regulated by brain ischemia using a novel SUMO transgenic mouse. METHODS: To enable SUMO proteomics analysis in vivo, we generated transgenic mice conditionally expressing tagged SUMO1-3 paralogues. Transgenic mice were subjected to 10 minutes forebrain ischemia and 1 hour of reperfusion. SUMO3-conjugated proteins were enriched by anti-FLAG affinity purification and analyzed by liquid chromatography-tandem mass spectrometry. RESULTS: Characterization of SUMO transgenic mice demonstrated that all 3 tagged SUMO paralogues were functionally active, and expression of exogenous SUMOs did not modify the endogenous SUMOylation machinery. Proteomics analysis identified 112 putative SUMO3 substrates of which 91 candidates were more abundant in the ischemia group than the sham group. Data analysis revealed processes/pathways with putative neuroprotective functions, including glucocorticoid receptor signaling, RNA processing, and SUMOylation-dependent ubiquitin conjugation. CONCLUSIONS: The identified proteins/pathways modulated by SUMOylation could be the key to understand the mechanisms linking SUMOylation to neuroprotection, and thus provide new promising targets for therapeutic interventions. The new transgenic mouse will be an invaluable platform for analyzing the SUMO-modified proteome in models of human disorders and thereby help to mechanistically link SUMOylation to the pathological processes.


Subject(s)
Brain Ischemia/physiopathology , Ischemic Attack, Transient/physiopathology , Stroke/physiopathology , Ubiquitins/genetics , Ubiquitins/metabolism , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/metabolism , Mass Spectrometry , Mice , Mice, Transgenic , Proteome/genetics , Proteome/metabolism , Proteomics , RNA Processing, Post-Transcriptional , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Stroke/genetics , Stroke/metabolism
20.
J Cereb Blood Flow Metab ; 34(3): 425-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24301296

ABSTRACT

Ubiquitylation is a posttranslational protein modification that modulates various cellular processes of key significance, including protein degradation and DNA damage repair. In animals subjected to transient cerebral ischemia, ubiquitin-conjugated proteins accumulate in Triton-insoluble aggregates. Although this process is widely considered to modulate the fate of postischemic neurons, few attempts have been made to characterize the ubiquitin-modified proteome in these aggregates. We performed proteomics analyses to identify ubiquitylated proteins in postischemic aggregates. Mice were subjected to 10 minutes of forebrain ischemia and 4 hours of reperfusion. The hippocampi were dissected, aggregates were isolated, and trypsin-digested after spiking with GG-BSA as internal standard. K-ɛ-GG-containing peptides were immunoprecipitated and analyzed by label-free quantitative liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. We identified 1,664 peptides to 520 proteins containing at least one K-ɛ-GG. Sixty-six proteins were highly ubiquitylated, with 10 or more K-ɛ-GG peptides. Based on selection criteria of greater than fivefold increase and P<0.001, 763 peptides to 272 proteins were highly enriched in postischemic aggregates. These included proteins involved in important neuronal functions and signaling pathways that are impaired after ischemia. Results of this study could serve as an important platform to uncover the mechanisms linking insoluble ubiquitin aggregates to the functions of postischemic neurons.


Subject(s)
Ischemic Attack, Transient/metabolism , Prosencephalon/metabolism , Proteome/metabolism , Ubiquitin/metabolism , Ubiquitinated Proteins/metabolism , Animals , Blotting, Western , Chromatography, Liquid , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Peptide Fragments/metabolism , Prosencephalon/blood supply , Proteomics , Tandem Mass Spectrometry , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...