Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Biol Chem ; 288(12): 8491-8504, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23382383

ABSTRACT

Apoptosis can occur in the myocardium under a variety of pathological conditions, including myocardial infarction and heart failure. The forkhead family of transcription factor Foxo3a plays a pivotal role in apoptosis; however, its role in regulating cardiac apoptosis remains to be fully elucidated. We showed that enforced expression of Foxo3a inhibits cardiomyocyte apoptosis, whereas knockdown of endogenous Foxo3a sensitizes cardiomyocytes to undergo apoptosis. The apoptosis repressor with caspase recruitment domain (ARC) is a potent anti-apoptotic protein. Here, we demonstrate that it attenuates the release of calcium from the sarcoplasmic reticulum and inhibits calcium elevations in the cytoplasm and mitochondria provoked by oxidative stress in cardiomyocytes. Furthermore, Foxo3a is shown to maintain cytoplasmic and mitochondrial calcium homeostasis through ARC. We observed that Foxo3a knock-out mice exhibited enlarged myocardial infarction sizes upon ischemia/reperfusion, and ARC transgenic mice demonstrated reduced myocardial infarction and balanced calcium levels in mitochondria and sarcoplasmic reticulum. Moreover, we showed that Foxo3a activates ARC expression by directly binding to its promoter. This study reveals that Foxo3a maintains calcium homeostasis and inhibits cardiac apoptosis through trans-activation of the ARC promoter. These findings provided novel evidence that Foxo3a and ARC constitute an anti-apoptotic pathway that regulates calcium homeostasis in the heart.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Calcium Signaling , Calcium/metabolism , Forkhead Transcription Factors/physiology , Muscle Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Base Sequence , Caspase 3/metabolism , Cells, Cultured , Enzyme Activation , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Genes, Reporter , Humans , Hydrogen Peroxide/pharmacology , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Mitochondria/metabolism , Muscle Proteins/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Oxidants/pharmacology , Promoter Regions, Genetic , Rats , Rats, Wistar , Sarcoplasmic Reticulum/metabolism , Transcriptional Activation
2.
PLoS One ; 16(7): e0254943, 2021.
Article in English | MEDLINE | ID: mdl-34310645

ABSTRACT

OBJECTIVE: Although myriad researches upon the associations between LncRNA H19 polymorphic variants (rs2839698 G>A, rs217727 G>A, rs2107425 C>T, rs2735971 A>G and rs3024270 C>G) and the susceptibility to cancer have been conducted, these results remained contradictory and perplexing. Basing on that, a systematic review and updated meta-analysis was performed to anticipate a fairly precise assessment about such associations. METHODS: We retrieved the electronic databases EMBASE, PubMed and Web of Science for valuable academic studies before February 28, 2021. Ultimately, 28 of which were encompassed after screening in this meta-analysis, and the available data was extracted and integrated. The pooled odds ratios (ORs) with 95% confidence intervals (CIs) was used to evaluate such associations. For multi-level investigation, subgroup analysis derived from source of controls together with genotypic method was preformed. RESULTS: Eventually, 28 articles altogether embodying 57 studies were included in this meta-analysis. The results illuminated that LncRNA H19 polymorphisms mentioned above were all irrelevant to cancer susceptibility. Nevertheless, crucial results were found concentrated in population-based control group when subgroup analysis by source of controls were performed in H19 mutation rs2839698 and rs2735971. Meanwhile, in the stratification analysis by genotypic method, apparent cancer risks were discovered by TaqMan method in H19 mutation rs2107425 and rs3024270. Then, trial sequential analysis demonstrated that the results about such associations were firm evidence of effect. CONCLUSION: Therefore, this meta-analysis indicated that LncRNA H19 polymorphisms were not associated with the susceptibility to human cancer. However, after the stratification analysis, inconsistent results still existed in different genotypic method and source of control. Thus, more high-quality studies on cancer patients of different factors were needed to confirm these findings.


Subject(s)
Genetic Predisposition to Disease , Neoplasms/pathology , RNA, Long Noncoding/genetics , Case-Control Studies , Databases, Factual , Humans , Neoplasms/genetics , Odds Ratio , Polymorphism, Single Nucleotide , RNA, Long Noncoding/metabolism
3.
J Clin Invest ; 129(11): 4593-4608, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31545295

ABSTRACT

The majority of patients with diabetic macular edema (DME), the most common cause of vision loss in working-age Americans, do not respond adequately to current therapies targeting VEGFA. Here, we show that expression of angiopoietin-like 4 (ANGPTL4), a HIF-1-regulated gene product, is increased in the eyes of diabetic mice and patients with DME. We observed that ANGPTL4 and VEGF act synergistically to destabilize the retinal vascular barrier. Interestingly, while ANGPTL4 modestly enhanced tyrosine phosphorylation of VEGF receptor 2, promotion of vascular permeability by ANGPTL4 was independent of this receptor. Instead, we found that ANGPTL4 binds directly to neuropilin 1 (NRP1) and NRP2 on endothelial cells (ECs), leading to rapid activation of the RhoA/ROCK signaling pathway and breakdown of EC-EC junctions. Treatment with a soluble fragment of NRP1 (sNRP1) prevented ANGPTL4 from binding to NRP1 and blocked ANGPTL4-induced activation of RhoA as well as EC permeability in vitro and retinal vascular leakage in diabetic animals in vivo. In addition, sNRP1 reduced the stimulation of EC permeability by aqueous fluid from patients with DME. Collectively, these data identify the ANGPTL4/NRP/RhoA pathway as a therapeutic target for the treatment of DME.


Subject(s)
Angiopoietin-Like Protein 4/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Macular Edema/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Macular Edema/pathology , Mice , Neuropilin-1/metabolism , Neuropilin-2/metabolism , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , rhoA GTP-Binding Protein/metabolism
4.
Cell Death Differ ; 26(9): 1600-1614, 2019 09.
Article in English | MEDLINE | ID: mdl-30390091

ABSTRACT

Mitochondrial superoxide dismutase 2 (SOD2) is a major antioxidant defense enzyme. Here we provide evidence that SOD2 plays critical roles in maintaining calcium homeostasis in newly generated embryonic cerebral cortical neurons, which is essential for normal mitochondrial function and subcellular distribution, and neurite outgrowth. Primary cortical neurons in cultures established from embryonic day 15 SOD2+/+ and SOD2-/- mice appear similar during the first 24 h in culture. During the ensuing two days in culture, SOD2-/- neurons exhibit a profound reduction of neurite outgrowth and their mitochondria become fragmented and accumulate in the cell body. The structural abnormalities of the mitochondria are associated with reduced levels of phosphorylated (S637) dynamin related protein 1 (Drp1), a major mitochondrial fission-regulating protein, whereas mitochondrial fusion regulating proteins (OPA1 and MFN2) are relatively unaffected. Mitochondrial fission and Drp1 dephosphorylation coincide with impaired mitochondrial Ca2+ buffering capacity and an elevation of cytosolic Ca2+ levels. Treatment of SOD2-/- neurons with the Ca2+ chelator BAPTA-AM significantly increases levels of phosphorylated Drp1, reduces mitochondrial fragmentation and enables neurite outgrowth.


Subject(s)
Cerebral Cortex/growth & development , Dynamins/genetics , Neurons/metabolism , Superoxide Dismutase/genetics , Animals , Calcium/metabolism , Cerebral Cortex/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Neuronal Outgrowth/genetics , Phosphorylation/genetics
5.
Nat Commun ; 10(1): 1886, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015456

ABSTRACT

Intermittent food deprivation (fasting, IF) improves mood and cognition and protects neurons against excitotoxic degeneration in animal models of epilepsy and Alzheimer's disease (AD). The mechanisms by which neuronal networks adapt to IF and how such adaptations impact neuropathological processes are unknown. We show that hippocampal neuronal networks adapt to IF by enhancing GABAergic tone, which is associated with reduced anxiety-like behaviors and improved hippocampus-dependent memory. These neuronal network and behavioral adaptations require the mitochondrial protein deacetylase SIRT3 as they are abolished in SIRT3-deficient mice and wild type mice in which SIRT3 is selectively depleted from hippocampal neurons. In the AppNL-G-F mouse model of AD, IF reduces neuronal network hyperexcitability and ameliorates deficits in hippocampal synaptic plasticity in a SIRT3-dependent manner. These findings demonstrate a role for a mitochondrial protein deacetylase in hippocampal neurons in behavioral and GABAergic synaptic adaptations to IF.


Subject(s)
Alzheimer Disease/diet therapy , Fasting/physiology , GABAergic Neurons/metabolism , Hippocampus/physiology , Sirtuin 3/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/physiology , Cognition/physiology , Cortical Excitability/physiology , Disease Models, Animal , Hippocampus/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Nerve Net/physiology , Neuronal Plasticity/physiology , Oxidative Stress/physiology , Sirtuin 3/genetics , Superoxide Dismutase/genetics
6.
Cell Metab ; 23(1): 128-42, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26698917

ABSTRACT

The impact of mitochondrial protein acetylation status on neuronal function and vulnerability to neurological disorders is unknown. Here we show that the mitochondrial protein deacetylase SIRT3 mediates adaptive responses of neurons to bioenergetic, oxidative, and excitatory stress. Cortical neurons lacking SIRT3 exhibit heightened sensitivity to glutamate-induced calcium overload and excitotoxicity and oxidative and mitochondrial stress; AAV-mediated Sirt3 gene delivery restores neuronal stress resistance. In models relevant to Huntington's disease and epilepsy, Sirt3(-/-) mice exhibit increased vulnerability of striatal and hippocampal neurons, respectively. SIRT3 deficiency results in hyperacetylation of several mitochondrial proteins, including superoxide dismutase 2 and cyclophilin D. Running wheel exercise increases the expression of Sirt3 in hippocampal neurons, which is mediated by excitatory glutamatergic neurotransmission and is essential for mitochondrial protein acetylation homeostasis and the neuroprotective effects of running. Our findings suggest that SIRT3 plays pivotal roles in adaptive responses of neurons to physiological challenges and resistance to degeneration.


Subject(s)
Mitochondria/enzymology , Neurons/physiology , Sirtuin 3/physiology , Acetylation , Adaptation, Physiological , Animals , Calcium/metabolism , Cells, Cultured , Energy Metabolism , Hippocampus/cytology , Membrane Potential, Mitochondrial , Mice, Knockout , Mitochondrial Proteins/metabolism , Neostriatum/cytology , Nerve Degeneration/enzymology , Physical Conditioning, Animal , Protective Factors , Protein Processing, Post-Translational , Running/physiology , Stress, Physiological
7.
Yi Chuan ; 27(4): 589-94, 2005 Jul.
Article in Zh | MEDLINE | ID: mdl-16120584

ABSTRACT

Using in situ hybridization on tissue sections with DIG antisense RNA probe, the distribution of zebrafish (Danio rerio) nanos1 mRNA during oogenesis and spermatogenesis was detected, and the expression and function of nanos 1 during zebrafish gametogenesis was preliminarily studied. The results were as follows: nanos 1 mRNA uniformly dispersed throughout the cytoplasm of oocytes at all stages. Strong expression of nanos1 mRNA was observed in oogonia and stage I, II oocytes, but the signal became weaker in later stage oocytes. Strong expression of nanos 1 mRNA was detected in spermatogonia and relatively weaker signal was found in primary spermatocytes but no nanos 1 mRNA expression was observed in spermatids. The results suggested that nanos 1 may play an important role in maintenance and functioning of the germline stem cell-oogonia and spermatogonia.


Subject(s)
Gametogenesis/genetics , Gene Expression Regulation, Developmental , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Female , In Situ Hybridization , Male , Oocytes/metabolism , Oogonia/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins , Spermatogonia/metabolism , Zebrafish/growth & development
8.
Mol Cell Biol ; 35(7): 1081-96, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605334

ABSTRACT

Insults to nuclear DNA induce multiple response pathways to mitigate the deleterious effects of damage and mediate effective DNA repair. G-protein-coupled receptor kinase-interacting protein 2 (GIT2) regulates receptor internalization, focal adhesion dynamics, cell migration, and responses to oxidative stress. Here we demonstrate that GIT2 coordinates the levels of proteins in the DNA damage response (DDR). Cellular sensitivity to irradiation-induced DNA damage was highly associated with GIT2 expression levels. GIT2 is phosphorylated by ATM kinase and forms complexes with multiple DDR-associated factors in response to DNA damage. The targeting of GIT2 to DNA double-strand breaks was rapid and, in part, dependent upon the presence of H2AX, ATM, and MRE11 but was independent of MDC1 and RNF8. GIT2 likely promotes DNA repair through multiple mechanisms, including stabilization of BRCA1 in repair complexes; upregulation of repair proteins, including HMGN1 and RFC1; and regulation of poly(ADP-ribose) polymerase activity. Furthermore, GIT2-knockout mice demonstrated a greater susceptibility to DNA damage than their wild-type littermates. These results suggest that GIT2 plays an important role in MRE11/ATM/H2AX-mediated DNA damage responses.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , DNA Repair , GTPase-Activating Proteins/metabolism , Phosphoproteins/metabolism , Animals , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , GTPase-Activating Proteins/analysis , GTPase-Activating Proteins/genetics , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phosphoproteins/analysis , Phosphoproteins/genetics
9.
Int J Endocrinol ; 2012: 739428, 2012.
Article in English | MEDLINE | ID: mdl-22754566

ABSTRACT

A strong connection between neuronal and metabolic health has been revealed in recent years. It appears that both normal and pathophysiological aging, as well as neurodegenerative disorders, are all profoundly influenced by this "neurometabolic" interface, that is, communication between the brain and metabolic organs. An important aspect of this "neurometabolic" axis that needs to be investigated involves an elucidation of molecular factors that knit these two functional signaling domains, neuronal and metabolic, together. This paper attempts to identify and discuss a potential keystone signaling factor in this "neurometabolic" axis, that is, the epidermal growth factor receptor (EGFR). The EGFR has been previously demonstrated to act as a signaling nexus for many ligand signaling modalities and cellular stressors, for example, radiation and oxidative radicals, linked to aging and degeneration. The EGFR is expressed in a wide variety of cells/tissues that pertain to the coordinated regulation of neurometabolic activity. EGFR signaling has been highlighted directly or indirectly in a spectrum of neurometabolic conditions, for example, metabolic syndrome, diabetes, Alzheimer's disease, cancer, and cardiorespiratory function. Understanding the positioning of the EGFR within the neurometabolic domain will enhance our appreciation of the ability of this receptor system to underpin highly complex physiological paradigms such as aging and neurodegeneration.

10.
PLoS One ; 7(5): e36911, 2012.
Article in English | MEDLINE | ID: mdl-22606307

ABSTRACT

As pharmacological data sets become increasingly large and complex, new visual analysis and filtering programs are needed to aid their appreciation. One of the most commonly used methods for visualizing biological data is the Venn diagram. Currently used Venn analysis software often presents multiple problems to biological scientists, in that only a limited number of simultaneous data sets can be analyzed. An improved appreciation of the connectivity between multiple, highly-complex datasets is crucial for the next generation of data analysis of genomic and proteomic data streams. We describe the development of VENNTURE, a program that facilitates visualization of up to six datasets in a user-friendly manner. This program includes versatile output features, where grouped data points can be easily exported into a spreadsheet. To demonstrate its unique experimental utility we applied VENNTURE to a highly complex parallel paradigm, i.e. comparison of multiple G protein-coupled receptor drug dose phosphoproteomic data, in multiple cellular physiological contexts. VENNTURE was able to reliably and simply dissect six complex data sets into easily identifiable groups for straightforward analysis and data output. Applied to complex pharmacological datasets, VENNTURE's improved features and ease of analysis are much improved over currently available Venn diagram programs. VENNTURE enabled the delineation of highly complex patterns of dose-dependent G protein-coupled receptor activity and its dependence on physiological cellular contexts. This study highlights the potential for such a program in fields such as pharmacology, genomics, and bioinformatics.


Subject(s)
Databases, Factual/statistics & numerical data , Pharmacology/statistics & numerical data , Software , Cell Line, Tumor , Computational Biology , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Humans , Ligands , Methacholine Chloride/administration & dosage , Muscarinic Agonists/administration & dosage , Phosphorylation , Proteomics/statistics & numerical data , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry
12.
Mol Cell Biol ; 28(2): 564-74, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17998337

ABSTRACT

p53 plays an important role in regulating apoptosis. However, the molecular mechanism by which it initiates the apoptotic program still remains to be fully understood. Here, we report that p53 can transcriptionally target the antiapoptotic protein, apoptosis repressor with caspase recruitment domain (ARC). Our results show that reactive oxygen species and anoxia lead to the up-regulation of p53 expression. Concomitantly, ARC is down-regulated at both the protein and mRNA levels. Knockdown of p53 expression can attenuate the decreases in ARC protein and mRNA levels, indicating that ARC down-regulation is a consequence of p53 activation. Strikingly, p53-induced ARC repression occurs in a transcription-dependent manner. We further demonstrate that the p53 up-regulated modulator of apoptosis (PUMA) and Bad are up-regulated in response to the stimulation with reactive oxygen species or anoxia, and p53 is responsible for their up-regulation. ARC can interact with PUMA or Bad via its N terminus. Such an interaction displaces the association of PUMA or Bad with Bcl-2. ARC repression by p53 leads to its failure to counteract the proapoptotic activity of PUMA and Bad. Thus, our data reveal a novel p53 apoptotic pathway in which it initiates apoptosis by transcriptionally repressing ARC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Muscle Proteins/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Binding Sites , Caspase 8/metabolism , Cell Line , Daunorubicin/pharmacology , Down-Regulation/drug effects , Enzyme Activation , Humans , Hydrogen Peroxide/pharmacology , Muscle Proteins/genetics , Oxygen/pharmacology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL