Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 9: 1984, 2018.
Article in English | MEDLINE | ID: mdl-30279688

ABSTRACT

Bile acids (BAs) control metabolism and inflammation by interacting with several receptors. Here, we report that intravenous infusion of taurodeoxycholate (TDCA) decreases serum pro-inflammatory cytokines, normalizes hypotension, protects against renal injury, and prolongs mouse survival during sepsis. TDCA increases the number of granulocytic myeloid-derived suppressor cells (MDSCLT) distinctive from MDSCs obtained without TDCA treatment (MDSCL) in the spleen of septic mice. FACS-sorted MDSCLT cells suppress T-cell proliferation and confer protection against sepsis when adoptively transferred better than MDSCL. Proteogenomic analysis indicated that TDCA controls chromatin silencing, alternative splicing, and translation of the immune proteome of MDSCLT, which increases the expression of anti-inflammatory molecules such as oncostatin, lactoferrin and CD244. TDCA also decreases the expression of pro-inflammatory molecules such as neutrophil elastase. These findings suggest that TDCA globally edits the proteome to increase the number of MDSCLT cells and affect their immune-regulatory functions to resolve systemic inflammation during sepsis.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Sepsis/immunology , T-Lymphocytes/immunology , Taurodeoxycholic Acid/metabolism , Animals , Cell Count , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Immune Tolerance , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncostatin M/genetics , Oncostatin M/metabolism
2.
Cell Mol Life Sci ; 70(4): 711-28, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23052207

ABSTRACT

Argonaute 2 (Ago2) is a pivotal regulator of cell fate in adult stem cells. Its expression is significantly downregulated in late passages of cells, concomitant with a prominent increase in Ago2 cytosolic localization in single cells. Nuclear localization of Ago2 is crucial for the survival, proliferation, and differentiation of hATSCs (human adipose tissue-derived stem cells), mediated by the specific binding of the regulatory regions of functional genes, which positively or negatively altered gene expression. Ago2 targets genes that control stemness, reactive oxygen species scavenging, and microRNA expression, all of which are crucial for hATSC survival and self-renewal. Ago2 promotes cell proliferation and self-renewal by activating the expression of octamer-binding transcription factor 4 (Oct4). We confirmed the direct regulation of Oct4 activity by Ago2, as indicated by the results of the ChIP analysis. Methyl-CpG-binding protein 6 (MBD6) was detected as an Oct4 regulatory gene. As predicted, knockdown of MBD6 expression attenuated cell proliferation and eventually induced cell death. We hypothesized that MBD6 functions downstream of Oct4 in the regulation of stemness-related genes, cell proliferation, self-renewal activity, and survival. MBD6 also promoted cell transdifferentiation into neural and endodermal ß-cells while significantly attenuating differentiation into the mesodermal lineage. We demonstrate that MBD6 is regulated by Ago2 via an interaction with Oct4, which alters self-renewal and gene expression in hATSCs. MBD6 was promoted cell proliferation through a novel set of signal mediators that may influence differentiation by repressing MBD2 and MBD3, which are possibly recruited by germ cell nuclear factor (GCNF).


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/cytology , DNA-Binding Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Adult , Adult Stem Cells/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Multigene Family , Up-Regulation
3.
Brain ; 135(Pt 4): 1237-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22466292

ABSTRACT

MicroRNAs have been shown to effectively regulate gene expression at the translational level. Recently, we identified novel microRNAs that were upregulated in a mouse model of spinal cord injury. Among those, we have focused on microRNA 486, which directly represses NeuroD6 expression through a conserved sequence in its untranslated region. We correlated the overexpression of microRNA 486 in motor neurons with a poor outcome due to progressive neurodegeneration and a pathophysiology that is mediated by reactive oxygen species. The expression of microRNA 486 was induced by reactive oxygen species that were produced by inflammatory factors, and reactive oxygen species were accumulated in response to the knockdown of NeuroD6, which enhances the downregulation of glutathione peroxidase 3 and thioredoxin-like 1 after traumatic spinal cord injury. NeuroD6 directly bound to regulatory regions of thioredoxin-like 1 and glutathione peroxidase 3 in motor neurons and activated their expression, which promoted reactive oxygen species scavenging. Moreover, knocking down microRNA 486 induced the expression of NeuroD6, which effectively ameliorated the spinal cord injury and allowed the mice to recover motor function. The infusion of exogenic NeuroD6 in spinal cord injury lesions effectively blocked apoptosis by reactivating thioredoxin-like 1 and glutathione peroxidase 3, which was accompanied by a recovery of motor function. Collectively, these findings have identified a novel microRNA in spinal cord injury lesions called microRNA 486, demonstrating a new role for NeuroD6 in neuroprotection, and suggest a potential therapeutic target for spinal cord injuries.


Subject(s)
Gene Expression Regulation/physiology , MicroRNAs/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caspase 3/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred ICR , MicroRNAs/genetics , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/metabolism , Myelin Basic Protein/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurofilament Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Peroxidase/metabolism , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Recovery of Function/drug effects , Recovery of Function/genetics , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Time Factors
4.
Antioxid Redox Signal ; 16(5): 383-99, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21995449

ABSTRACT

AIMS: Argonaute2 (Ago2) plays a fundamental role in microRNA-mediated gene regulation through its intrinsic endonuclease activity. In this study we demonstrate the novel functions and molecular mechanisms by which nuclear Ago2 directly regulates HSP (heat shock protein) 60 expression and stem cell self-renewal. HSP60 is a crucial regulator of ROS (reactive oxygen species), senescence, and apoptotic cell death in several tissues and cell types. RESULTS: HSP60 is regulated via inactivation of p38/JNK and p53 and binds directly to the regulatory regions of the TERT, c-myc, GPx3, p53, and STAT3 genes. Using HSP60 CHIP-PCR experiments, we show that HSP60 binds directly to the Oct4 and Nanog genes and directly regulates Oct4 and other stemness genes involved in human adipose tissue-derived stem cell (hATSC) differentiation. HSP60 also positively regulates ROS-scavenging factors, including GPx3 and TXNL1, which directly modulate cytosolic ROS in hATSCs. Moreover, our study shows that Oct4 regulates HSP60 expression and controls hATSC survival and self-renewal after binding to the HSP60 gene. Furthermore, HSP60-mediated regulation of Oct4 contributes to neuronal and endodermal ß-cell differentiation of hATSCs in vitro and in vivo and downregulates mesoderm-specific gene expression. INNOVATION AND CONCLUSION: We show that increased levels of Ago2 or HSP60 effectively induce nuclear localization of HSP60, which directly controls Oct4, c-Myc, p53, TERT, and STAT3 for transdifferentiation programs. Collectively, we suggest a novel model in which nuclear Ago2 controls HSP60 in hATSCs.


Subject(s)
Argonaute Proteins/metabolism , Cell Nucleus/genetics , Chaperonin 60/metabolism , Octamer Transcription Factor-3/metabolism , Stem Cells/metabolism , Aging , Argonaute Proteins/genetics , Cell Death , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Chaperonin 60/genetics , Humans , Octamer Transcription Factor-3/genetics , Reactive Oxygen Species/metabolism , Stem Cells/cytology
5.
Antioxid Redox Signal ; 16(2): 95-111, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21902595

ABSTRACT

AIMS: Argonaute2 (Ago2) has intrinsic endonuclease activity in microRNA processing that plays a fundamental role in gene regulation. In this study, we demonstrate novel functions and molecular mechanisms of nuclear Ago2 in the self-renewal and plasticity of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs). RESULTS: Nuclear Ago2 binds to a set of regulatory genes, including Ago2 itself, Oct4, Sox2, Nanog, GATA, STAT3, and ß-catenin, that potentially target fundamental functions of stem cells. Direct regulation of the stemness genes by nuclear Ago2 was also crucial for cell self-renewal, survival, and differentiation into various types of tissues or cells, including neural cells and ß-cells. Moreover, regulation of Oct4 by Ago2 directly controls the stem cell plasticity-determining signal mediators JAK2/STAT3 and Wnt5A/ß-catenin and positively regulates cell proliferation and differentiation via blockage of ROS generation and P38/JNK inactivation. Nuclear Ago2 or stemness expression lead increased stem cell identity and decreased differentiation into a mesodermal lineage but also led to increased neural differentiation and ß-cell differentiation in hUCB-MSCs. Nuclear Ago2-mediated stemness expression in hUCB-MSCs is also involved in cell survival, helping cells escape apoptotic cell death via inactivation of P38/JNK, caspase-3, and Bax. INNOVATION AND CONCLUSION: This study reveals that nuclear Ago2 globally controls stem cell self-renewal and differentiation through direct regulation of stemness genes and important signal mediator activation following inactivation of ROS/P38/JNK and activation of the JAK/STAT3 and Wnt/ ß-catenin signal pathways.


Subject(s)
Argonaute Proteins/physiology , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Apoptosis , Argonaute Proteins/genetics , Base Sequence , Humans , Janus Kinases/metabolism , Molecular Sequence Data , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , Wnt Proteins/metabolism , Wnt-5a Protein , beta Catenin/metabolism
6.
Hum Gene Ther ; 23(5): 508-20, 2012 May.
Article in English | MEDLINE | ID: mdl-22182208

ABSTRACT

MicroRNAs (miRNAs) compose a relatively new discipline in biomedical research, and many physiological processes in disease have been associated with changes in miRNA expression. Several studies report that miRNAs participate in biological processes such as the control of secondary injury in several disease models. Recently, we identified novel miRNAs that were abnormally up-regulated in a traumatic spinal cord injury (SCI). In the current study, we focused on miR20a, which causes continuing motor neuron degeneration when overexpressed in SCI lesions. Blocking miR20a in SCI animals led to neural cell survival and eventual neurogenesis with rescued expression of the key target gene, neurogenin 1 (Ngn1). Infusion of siNgn1 resulted in functional deficit in the hindlimbs caused by aggressive secondary injury and actively enhanced the inflammation involved in secondary injury progression. The events involving miR20a underlie motor neuron and myelin destruction and pathophysiology and ultimately block regeneration in injured spinal cords. Inhibition of miR20a expression effectively induced definitive motor neuron survival and neurogenesis, and SCI animals showed improved functional deficit. In this study, we showed that abnormal expression of miR20a induces secondary injury, which suggests that miR20a could be a potential target for therapeutic intervention following SCI.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , MicroRNAs/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , RNA Interference , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/administration & dosage , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Hindlimb/pathology , Humans , Mice , MicroRNAs/genetics , Motor Neurons/metabolism , Motor Neurons/pathology , Myelin Sheath/pathology , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/antagonists & inhibitors , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
7.
Antioxid Redox Signal ; 16(10): 1046-60, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22149086

ABSTRACT

AIMS: Neuropathic pain is a well-known type of chronic pain caused by damage to the nervous system. Until recently, researchers have been primarily focused on identifying the cellular or chemical sources of neuropathic pain or have approached neuropathic pain via the basis of biological study. We investigated whether mmu-mir-23b (miR23b) infusion can alleviate pain by compensating for the abnormally downregulated miR23b by reducing the expression of its target gene, NADPH oxidase 4 (NOX4), a reactive oxygen species (ROS) family member overexpressed in neuropathic pain. RESULTS: Ectopic miR23b expression effectively downregulated NOX4 and was normalized to GAD65/67 expression. Moreover, the animals with neuropathic pain showed significant improvements in the paw withdrawal thresholds following miR23b infusion. Normalizing miR23b expression in tissue lesions caused by neuropathic pain induction reduced inflammatory mediator expression and increased the level of several ROS scavengers. Moreover, GABAergic neurons coexpressed suboptimal levels of miR23b and elevated NOX4/ROS after pain induction at the cellular level. MiR23b protects GABAergic neurons against ROS/p38/JNK-mediated apoptotic death. By evaluating the functional behavior of the mice receiving pain/miR23b, normal/anti-miR23b, or anti-miR23b/si-NOX4, the positive role of miR23b and the negative role of NOX4 in neuropathic pain were confirmed. INNOVATION AND CONCLUSION: Based on this study, we conclude that miR23b plays a crucial role in the amelioration of neuropathic pain in the injured spinal cord by inactivating its target gene, NOX4, and protecting GABAergic neurons from cell death. We finally suggest that miR23b may provide attractive diagnostic and therapeutic resources for effective pain modulation in neuropathic pain.


Subject(s)
Gene Silencing , MicroRNAs/metabolism , NADPH Oxidases/genetics , Neuralgia/genetics , Spinal Cord/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Female , Mice , MicroRNAs/administration & dosage , MicroRNAs/genetics , NADPH Oxidase 4 , Neuralgia/therapy
8.
Hum Gene Ther ; 22(12): 1551-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21810014

ABSTRACT

In the present study, we show that Rex-1 mRNA and protein are found at high levels in both 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU)-resistant glioma cell subpopulations and malignant glioblastoma multiforme (GBM) tissue. We used a combination therapy of small interfering RNA (siRNA) against Rex-1 (siRex-1) and BCNU to target GBM cells. Rex-1 siRNA/BCNU treatment resulted in growth inhibition and a diminished S phase. The treatment efficiently induced P38/JNK and Akt/PI3K/GSK3ß signaling and led to apoptosis both in vitro and in vivo. We also show that Rex-1/ABCG2 (ATP binding cassette transporter G2)-coexpressing subpopulations were chemoresistant; however, BCNU was not a substrate for ABCG2. siRex-1 treatment led to cell death in GBM subpopulations by promoting apoptosis. Moreover, siRex-1/BCNU combination therapy targeted both the major population and cancer stem cell-like subpopulations. Our findings are important for the development of clinical applications to treat GBM.


Subject(s)
Apoptosis , Carmustine/therapeutic use , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Glioma/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , RNA, Small Interfering/genetics , Animals , Antineoplastic Agents/therapeutic use , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Methylation , Female , Glioma/genetics , Humans , Immunoenzyme Techniques , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Aging Cell ; 10(2): 277-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21241449

ABSTRACT

Argonaute 2 (Ago2) has a leading function in miRNA-induced RNA silencing, a conserved gene regulatory mechanism in cells and organisms. miRNAs are critical for stem cell self-renewal, development, and other functions. Here, we report that nuclear Ago2, by binding to a specific region of functional genes, directly controls adipose tissue-derived stem cell (ATSC) survival in response to a critical dose of reactive oxygen species (ROS)-mediated oxidative cell damage or senescence. The role of nuclear Ago2 has not been previously reported. Here, we show that human ATSCs in which Ago2 was downregulated underwent apoptosis. Silencing of Ago2 in ATSCs significantly induces upregulation of miR10b and miR23b expression. These miRNAs directly interfere with ROS-scavenging gene expression, such as TXNL1 and GPX3. Upregulation of miR10b and miR23b is sufficient to induce ATSC cell apoptosis via p38 MAPK phosphorylation and caspase 3 activation. In addition, Ago2 overexpression or interference by miR10b and miR23b expression in ATSCs partially rescued H(2) O(2) /ROS-mediated apoptotic cell death by upregulating the expression of TXNL2, JUNK, caspase-3, and cytochrome C. Nuclear Ago2-mediated miR10b and miR23b downregulation also allows cells to escape senescence, which results in telomerase reverse transcriptase, stemness overexpression, and improved self-renewal and differentiation through Wnt5a/ß-catenin activation. Argonaute 2 expression is critical for stem cells to escape senescence by downregulating miR10b and miR23b. The Ago2-binding gene selenoprotein N1 (SEPN1) was also effectively involved in ATSC survival and self-renewal through ROS-mediated p38 MAPK inactivation.


Subject(s)
Adipose Tissue/cytology , Cell Nucleus/metabolism , Cell Survival/physiology , Eukaryotic Initiation Factor-2/metabolism , Membrane Glycoproteins/metabolism , Muscle Proteins/metabolism , Receptors, Immunologic/metabolism , Selenoproteins/metabolism , Stem Cells/physiology , Apoptosis/physiology , Argonaute Proteins , Base Sequence , Cell Proliferation , Cells, Cultured , Eukaryotic Initiation Factor-2/genetics , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Muscle Proteins/genetics , Reactive Oxygen Species/metabolism , Receptors, Immunologic/genetics , Selenoproteins/genetics , Stem Cells/cytology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...