Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673852

ABSTRACT

One of the challenges of the mature nervous system is to maintain the stability of neural networks while providing a degree of plasticity to generate experience-dependent modifications. This plasticity-stability dynamism is regulated by perineuronal nets (PNNs) and is crucial for the proper functioning of the system. Previously, we found a relation between spinal PNNs reduction and maladaptive plasticity after spinal cord injury (SCI), which was attenuated by maintaining PNNs with activity-dependent therapies. Moreover, transgenic mice lacking the cartilage link protein 1 (Crtl1 KO mice) showed aberrant spinal PNNs and increased spinal plasticity. Therefore, the aim of this study is to evaluate the role of link protein 1 in the activity-dependent modulation of spinal PNNs surrounding motoneurons and its impact on the maladaptive plasticity observed following SCI. We first studied the activity-dependent modulation of spinal PNNs using a voluntary wheel-running protocol. This training protocol increased spinal PNNs in WT mice but did not modify PNN components in Crtl1 KO mice, suggesting that link protein 1 mediates the activity-dependent modulation of PNNs. Secondly, a thoracic SCI was performed, and functional outcomes were evaluated for 35 days. Interestingly, hyperreflexia and hyperalgesia found at the end of the experiment in WT-injured mice were already present at basal levels in Crtl1 KO mice and remained unchanged after the injury. These findings demonstrated that link protein 1 plays a dual role in the correct formation and in activity-dependent modulation of PNNs, turning it into an essential element for the proper function of PNN in spinal circuits.


Subject(s)
Extracellular Matrix Proteins , Mice, Knockout , Spinal Cord Injuries , Spinal Cord , Animals , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Mice , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Neuronal Plasticity , Motor Neurons/metabolism , Nerve Net/metabolism , Male , Proteoglycans/metabolism , Proteoglycans/genetics , Mice, Inbred C57BL
2.
Biomed Pharmacother ; 172: 116287, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38382328

ABSTRACT

Hypertension is the most prevalent modifiable risk factor for stroke and is associated with worse functional outcomes. Pharmacological inhibition of histone deacetylases by suberoylanilide hydroxamic acid (SAHA) modulates gene expression and has emerged as a promising therapeutic approach to reduce ischaemic brain injury. Here, we have tested the therapeutic potential of SAHA administered during reperfusion in adult male spontaneously hypertensive (SHR) rats subjected to transient middle cerebral artery occlusion (tMCAO; 90 min occlusion/24 h reperfusion). Animals received a single dose of SAHA (50 mg/kg) or vehicle i.p. at 1, 4, or 6 h after reperfusion onset. The time-course of brain histone H3 acetylation was studied. After tMCAO, drug brain penetrance and beneficial effects on behavioural outcomes, infarct volume, oedema, angiogenesis, blood-brain barrier integrity, cerebral artery oxidative stress and remodelling, and brain and vascular inflammation were evaluated. SAHA increased brain histone H3 acetylation from 1 to 6 h after injection, reaching the ischaemic brain administered during reperfusion. Treatment given at 4 h after reperfusion onset improved neurological score, reduced infarct volume and oedema, attenuated microglial activation, prevented exacerbated MCA angiogenic sprouting and blood-brain barrier breakdown, normalised MCA oxidative stress and remodelling, and modulated brain and cerebrovascular cytokine expression. Overall, we demonstrate that SAHA administered during early reperfusion exerts robust brain and vascular protection after tMCAO in hypertensive rats. These findings are aligned with previous research in ischaemic normotensive mice and help pave the way to optimise the design of clinical trials assessing the effectiveness and safety of SAHA in ischaemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Male , Rats , Animals , Mice , Vorinostat/pharmacology , Vorinostat/therapeutic use , Histone Deacetylases , Rats, Inbred SHR , Brain Ischemia/drug therapy , Histones , Stroke/drug therapy , Brain , Infarction , Edema
3.
Life Sci ; 336: 122286, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38007144

ABSTRACT

AIMS: Transient receptor potential vanilloid 2 (TRPV2) channels are expressed in both smooth muscle and endothelial cells and participate in vascular mechanotransduction and sensing of high temperatures and lipids. Nevertheless, the impact of TRPV2 channel activation by agonists on the coordinated and cell-type specific modulation of vasoreactivity is unknown. MAIN METHODS: Aorta from 2- to 4-months-old male Oncins France 1 mice was dissected and mounted in tissue baths for isometric tension measurements. TRPV2 channel expression was assessed by immunofluorescence and western blot in mice aortas and in cultured A7r5 rat aortic smooth muscle cells. KEY FINDINGS: TRPV2 channels were expressed in all three mouse aorta layers. Activation of TRPV2 channels with probenecid evoked endothelium-dependent relaxations through a mechanism that involved activation of smooth muscle Kir and Kv channels. In addition, TRPV2 channel inhibition with tranilast increased endothelium-independent relaxations to probenecid and this effect was abrogated by the KATP channel blocker glibenclamide, revealing that smooth muscle TRPV2 channels induce negative feedback on probenecid relaxations mediated via KATP channel inhibition. Exposure to the NO donor sodium nitroprusside increased TRPV2 channel translocation to the plasma membrane in cultured smooth muscle cells and enhanced negative feedback on probenecid relaxations. SIGNIFICANCE: In conclusion, we present the first evidence that TRPV2 channels may modulate vascular tone through a balance of opposed inputs from the endothelium and the smooth muscle leading to net vasodilation. The fact that TRPV2 channel-induced activity can be amplified by NO emphasizes the pathophysiological relevance of these findings.


Subject(s)
Endothelial Cells , Probenecid , Mice , Rats , Male , Animals , Probenecid/pharmacology , Mechanotransduction, Cellular , Aorta/metabolism , Vasodilation , Adenosine Triphosphate/metabolism , Endothelium, Vascular/physiology
5.
J Neurosci Res ; 100(6): 1331-1346, 2022 06.
Article in English | MEDLINE | ID: mdl-35218246

ABSTRACT

Peripheral nerve regeneration is limited after injury, especially in humans, due to the large distance the axons have to grow in the limbs. This process is highly dependent on the expression of neuroinflammatory factors produced by macrophages and glial cells. Given the importance of the epigenetic BET proteins on inflammation, we aimed to ascertain if BET inhibition may have an effect on axonal outgrowth. For this purpose, we treated female mice with JQ1 or vehicle after sciatic nerve crush injury and analyzed target reinnervation. We also used dorsal root ganglion (DRG) culture explants to analyze the effects of direct BET inhibition or treatment with conditioned medium from BET-inhibited macrophages. We observed that although JQ1 produced an enhancement of IL-4, IL-13, and GAP43 expression, it did not have an effect on sensory or motor reinnervation after crush injury in vivo. In contrast, JQ1 reduced neurite growth when interacting directly with DRG neurons ex vivo, whereas conditioned medium from JQ1-treated macrophages promoted neurite outgrowth. Therefore, BET-inhibited macrophages secrete pro-regenerative factors that induce neurite outgrowth, and that may counteract the direct inhibition of BET proteins in neurons in vivo. Finally, we observed an activation of the STAT6 pathway in DRG explants treated with conditioned medium from JQ1-treated macrophages. In conclusion, this study demonstrates that BET protein inhibition in macrophages provides a mechanism to enhance axonal outgrowth. However, specific targeting of BET proteins to macrophages will be needed to efficiently enhance functional recovery after nerve injury.


Subject(s)
Crush Injuries , Ganglia, Spinal , Animals , Cells, Cultured , Crush Injuries/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Female , Ganglia, Spinal/metabolism , Macrophages , Mice , Nerve Regeneration/physiology , Neurites , Neuronal Outgrowth
6.
J Pain ; 22(12): 1617-1630, 2021 12.
Article in English | MEDLINE | ID: mdl-34157407

ABSTRACT

Neuropathic pain is a common disability produced by enhanced neuronal excitability after nervous system injury. The pathophysiological changes that underlie the generation and maintenance of neuropathic pain require modifications of transcriptional programs. In particular, there is an induction of pro-inflammatory neuromodulators levels, and changes in the expression of ion channels and other factors intervening in the determination of the membrane potential in neuronal cells. We have previously found that inhibition of the BET proteins epigenetic readers reduced neuroinflammation after spinal cord injury. Within the present study we aimed to determine if BET protein inhibition may also affect neuroinflammation after a peripheral nerve injury, and if this would beneficially alter neuronal excitability and neuropathic pain. For this purpose, C57BL/6 female mice underwent spared nerve injury (SNI), and were treated with the BET inhibitor JQ1, or vehicle. Electrophysiological and algesimetry tests were performed on these mice. We also determined the effects of JQ1 treatment after injury on neuroinflammation, and the expression of neuronal components important for the maintenance of axon membrane potential. We found that treatment with JQ1 affected neuronal excitability and mechanical hyperalgesia after SNI in mice. BET protein inhibition regulated cytokine expression and reduced microglial reactivity after injury. In addition, JQ1 treatment altered the expression of SCN3A, SCN9A, KCNA1, KCNQ2, KCNQ3, HCN1 and HCN2 ion channels, as well as the expression of the Na+/K+ ATPase pump subunits. In conclusion, both, alteration of inflammation, and neuronal transcription, could be the responsible epigenetic mechanisms for the reduction of excitability and hyperalgesia observed after BET inhibition. Inhibition of BET proteins is a promising therapy for reducing neuropathic pain after neural injury. PERSPECTIVE: Neuropathic pain is a common disability produced by enhanced neuronal excitability after nervous system injury. The underlying pathophysiological changes require modifications of transcriptional programs. This study notes that inhibition of BET proteins is a promising therapy for reducing neuropathic pain after neural injury.


Subject(s)
Azepines/pharmacology , Hyperalgesia , Nerve Tissue Proteins/antagonists & inhibitors , Neuralgia , Neuroinflammatory Diseases , Peripheral Nerve Injuries , Receptors, Cell Surface/antagonists & inhibitors , Triazoles/pharmacology , Animals , Disease Models, Animal , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neuralgia/drug therapy , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/physiopathology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/physiopathology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology
7.
Nat Commun ; 10(1): 3028, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292434

ABSTRACT

Cerebellar neuronal progenitors undergo a series of divisions before irreversibly exiting the cell cycle and differentiating into neurons. Dysfunction of this process underlies many neurological diseases including ataxia and the most common pediatric brain tumor, medulloblastoma. To better define the pathways controlling the most abundant neuronal cells in the mammalian cerebellum, cerebellar granule cell progenitors (GCPs), we performed RNA-sequencing of GCPs exiting the cell cycle. Time-series modeling of GCP cell cycle exit identified downregulation of activity of the epigenetic reader protein Brd4. Brd4 binding to the Gli1 locus is controlled by Casein Kinase 1δ (CK1 δ)-dependent phosphorylation during GCP proliferation, and decreases during GCP cell cycle exit. Importantly, conditional deletion of Brd4 in vivo in the developing cerebellum induces cerebellar morphological deficits and ataxia. These studies define an essential role for Brd4 in cerebellar granule cell neurogenesis and are critical for designing clinical trials utilizing Brd4 inhibitors in neurological indications.


Subject(s)
Cerebellar Ataxia/genetics , Cerebellar Cortex/growth & development , Neural Stem Cells/physiology , Neurogenesis/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Casein Kinase Idelta , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cerebellar Ataxia/pathology , Cerebellar Cortex/cytology , Cerebellar Cortex/pathology , Disease Models, Animal , Down-Regulation , Humans , Mice , Mice, Knockout , Neurons/physiology , Nuclear Proteins/genetics , Phosphorylation/physiology , Primary Cell Culture , Transcription Factors/genetics , Zinc Finger Protein GLI1/metabolism
8.
J Neuroinflammation ; 16(1): 124, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31186006

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) usually causes a devastating lifelong disability for patients. After a traumatic lesion, disruption of the blood-spinal cord barrier induces the infiltration of macrophages into the lesion site and the activation of resident glial cells, which release cytokines and chemokines. These events result in a persistent inflammation, which has both detrimental and beneficial effects, but eventually limits functional recovery and contributes to the appearance of neuropathic pain. Bromodomain and extra-terminal domain (BET) proteins are epigenetic readers that regulate the expression of inflammatory genes by interacting with acetylated lysine residues. While BET inhibitors are a promising therapeutic strategy for cancer, little is known about their implication after SCI. Thus, the current study was aimed to investigate the anti-inflammatory role of BET inhibitors in this pathologic condition. METHODS: We evaluated the effectiveness of the BET inhibitor JQ1 to modify macrophage reactivity in vitro and to modulate inflammation in a SCI mice model. We analyzed the effects of BET inhibition in pro-inflammatory and anti-inflammatory cytokine production in vitro and in vivo. We determined the effectiveness of BET inhibition in tissue sparing, inflammation, neuronal protection, and behavioral outcome after SCI. RESULTS: We have found that the BET inhibitor JQ1 reduced the levels of pro-inflammatory mediators and increased the expression of anti-inflammatory cytokines. A prolonged treatment with JQ1 also decreased reactivity of microglia/macrophages, enhanced neuroprotection and functional recovery, and acutely reduced neuropathic pain after SCI. CONCLUSIONS: BET protein inhibition is an effective treatment to regulate cytokine production and promote neuroprotection after SCI. These novel results demonstrate for the first time that targeting BET proteins is an encouraging approach for SCI repair and a potential strategy to treat other inflammatory pathologies.


Subject(s)
Azepines/pharmacology , Cytokines/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Spinal Cord Injuries/metabolism , Triazoles/pharmacology , Animals , Cytokines/biosynthesis , Female , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Recovery of Function/drug effects
9.
Front Cell Neurosci ; 12: 158, 2018.
Article in English | MEDLINE | ID: mdl-29930500

ABSTRACT

Accumulating evidence suggests that epigenetic alterations lie behind the induction and maintenance of neuropathic pain. Neuropathic pain is usually a chronic condition caused by a lesion, or pathological change, within the nervous system. Neuropathic pain appears frequently after nerve and spinal cord injuries or diseases, producing a debilitation of the patient and a decrease of the quality of life. At the cellular level, neuropathic pain is the result of neuronal plasticity shaped by an increase in the sensitivity and excitability of sensory neurons of the central and peripheral nervous system. One of the mechanisms thought to contribute to hyperexcitability and therefore to the ontogeny of neuropathic pain is the altered expression, trafficking, and functioning of receptors and ion channels expressed by primary sensory neurons. Besides, neuronal and glial cells, such as microglia and astrocytes, together with blood borne macrophages, play a critical role in the induction and maintenance of neuropathic pain by releasing powerful neuromodulators such as pro-inflammatory cytokines and chemokines, which enhance neuronal excitability. Altered gene expression of neuronal receptors, ion channels, and pro-inflammatory cytokines and chemokines, have been associated to epigenetic adaptations of the injured tissue. Within this review, we discuss the involvement of these epigenetic changes, including histone modifications, DNA methylation, non-coding RNAs, and alteration of chromatin modifiers, that have been shown to trigger modification of nociception after neural lesions. In particular, the function on these processes of EZH2, JMJD3, MeCP2, several histone deacetylases (HDACs) and histone acetyl transferases (HATs), G9a, DNMT, REST and diverse non-coding RNAs, are described. Despite the effort on developing new therapies, current treatments have only produced limited relief of this pain in a portion of patients. Thus, the present review aims to contribute to find novel targets for chronic neuropathic pain treatment.

10.
Mol Cancer ; 17(1): 74, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29558959

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive malignant adult primary brain tumor. Despite surgical resection followed by radiotherapy and chemotherapy, the median survival rate is approximately 14 months. Although experimental therapies are in clinical trials for GBM, there is an urgent need for a peripheral GBM biomarker for measuring treatment response. As we have previously demonstrated that the long noncoding RNA HOX Transcript Antisense Intergenic RNA, or HOTAIR, is dysregulated in GBM and required for GBM cell proliferation, we hypothesized that HOTAIR expression may be utilized as a peripheral biomarker for GBM. HOTAIR expression was measured in serum from 43 GBM and 40 controls using quantitative real-time PCR (qRT-PCR). The PCR products were subsequently subcloned into pCR™4-TOPO®TA vectors for DNA sequencing. A ROC curve was also generated to examine HOTAIR's prognostic value. The amount of HOTAIR in serum exosomes and exosome-depleted supernatant was calculated by qRT-PCR. The relative HOTAIR expression was also investigated in 15 pairs of GBM serum and tumors. We detected HOTAIR in serum from GBM patients. HOTAIR levels in serum samples from GBM patients was significantly higher than in the corresponding controls (P < 0.0001). The area under the ROC curve distinguishing GBM patients from controls was 0.913 (95% CI: 0.845-0.982, P < 0.0001), with 86.1% sensitivity and 87.5% specificity at the cut-off value of 10.8. HOTAIR expression was significantly correlated with high grade brain tumors. In addition, Pearson correlation analysis indicated a medium correlation of serum HOTAIR levels and the corresponding tumor HOTAIR levels (r = 0.734, P < 0.01). We confirmed via sequencing that the amplified HOTAIR from serum contained the HOTAIR sequence and maps to the known HOTAIR locus at 12q13. The serum-derived exosomes contain HOTAIR and the purified exosomes were validated by western blot and nanoparticle tracking analysis. Importantly, our results demonstrate that serum HOTAIR can be used as a novel prognostic and diagnostic biomarker for GBM.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Cell-Free Nucleic Acids , Glioblastoma/diagnosis , Glioblastoma/genetics , RNA, Long Noncoding/genetics , Brain Neoplasms/blood , Brain Neoplasms/mortality , Exosomes , Glioblastoma/blood , Glioblastoma/mortality , Humans , Prognosis , RNA, Long Noncoding/blood , ROC Curve
12.
Proc Natl Acad Sci U S A ; 112(27): 8326-31, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26111795

ABSTRACT

Bromodomain and extraterminal (BET) domain proteins have emerged as promising therapeutic targets in glioblastoma and many other cancers. Small molecule inhibitors of BET bromodomain proteins reduce expression of several oncogenes required for Glioblastoma Multiforme (GBM) progression. However, the mechanism through which BET protein inhibition reduces GBM growth is not completely understood. Long noncoding RNAs (lncRNAs) are important epigenetic regulators with critical roles in cancer initiation and malignant progression, but mechanistic insight into their expression and regulation by BET bromodomain inhibitors remains elusive. In this study, we used Helicos single molecule sequencing to comprehensively profile lncRNAs differentially expressed in GBM, and we identified a subset of GBM-specific lncRNAs whose expression is regulated by BET proteins. Treatment of GBM cells with the BET bromdomain inhibitor I-BET151 reduced levels of the tumor-promoting lncRNA HOX transcript antisense RNA (HOTAIR) and restored the expression of several other GBM down-regulated lncRNAs. Conversely, overexpression of HOTAIR in conjunction with I-BET151 treatment abrogates the antiproliferative activity of the BET bromodomain inhibitor. Moreover, chromatin immunoprecipitation analysis demonstrated binding of Bromodomain Containing 4 (BRD4) to the HOTAIR promoter, suggesting that BET proteins can directly regulate lncRNA expression. Our data unravel a previously unappreciated mechanism through which BET proteins control tumor growth of glioblastoma cells and suggest that modulation of lncRNA networks may, in part, mediate the antiproliferative effects of many epigenetic inhibitors currently in clinical trials for cancer and other diseases.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation/genetics , Glioblastoma/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mice, Nude , Microscopy, Fluorescence , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , RNA, Long Noncoding/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Xenograft Model Antitumor Assays/methods
13.
Cell Rep ; 11(2): 249-60, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25843713

ABSTRACT

Although casein kinase 1δ (CK1δ) is at the center of multiple signaling pathways, its role in the expansion of CNS progenitor cells is unknown. Using mouse cerebellar granule cell progenitors (GCPs) as a model for brain neurogenesis, we demonstrate that the loss of CK1δ or treatment of GCPs with a highly selective small molecule inhibits GCP expansion. In contrast, CK1δ overexpression increases GCP proliferation. Thus, CK1δ appears to regulate GCP neurogenesis. CK1δ is targeted for proteolysis via the anaphase-promoting complex/cyclosome (APC/C(Cdh1)) ubiquitin ligase, and conditional deletion of the APC/C(Cdh1) activator Cdh1 in cerebellar GCPs results in higher levels of CK1δ. APC/C(Cdh1) also downregulates CK1δ during cell-cycle exit. Therefore, we conclude that APC/C(Cdh1) controls CK1δ levels to balance proliferation and cell-cycle exit in the developing CNS. Similar studies in medulloblastoma cells showed that CK1δ holds promise as a therapeutic target.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/biosynthesis , Casein Kinase Idelta/biosynthesis , Cdh1 Proteins/biosynthesis , Central Nervous System/growth & development , Neurogenesis/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Casein Kinase Idelta/genetics , Cdh1 Proteins/genetics , Cell Cycle/genetics , Cell Proliferation/genetics , Central Nervous System/metabolism , Cerebellum/growth & development , Cerebellum/metabolism , Gene Expression Regulation, Developmental , HeLa Cells , Humans , Mice , Neurons/metabolism , RNA Interference , Signal Transduction
14.
Cell Cycle ; 14(3): 417-24, 2015.
Article in English | MEDLINE | ID: mdl-25616418

ABSTRACT

Ubiquitin mediated proteolysis is required for transition from one cell cycle phase to another. For instance, the mitosis inhibitor Wee1 is targeted for degradation during S phase and G2 to allow mitotic entry. Wee1 is an essential tyrosine kinase required for the G2/M transition and S-phase progression. Although several studies have concentrated on Wee1 regulation during mitosis, few have elucidated its degradation during interphase. Our prior studies have demonstrated that Wee1 is degraded via CK1δ dependent phosphorylation during the S and G2/M phases of the cell cycle. Here we demonstrate that GSK3ß may work in concert with CK1δ to induce Wee1 destruction during interphase. We generated small molecules that specifically stabilized Wee1. We profiled these compounds against 296 kinases and found that they inhibit GSK3α and GSK3ß, suggesting that Wee1 may be targeted for proteolysis by GSK3. Consistent with this notion, known GSK3 inhibitors stabilized Wee1 and GSK3ß depletion reduced Wee1 turnover. Given Wee1's central role in cell cycle progression, we predicted that GSK3 inhibitors should limit cell proliferation. Indeed, we demonstrate that GSK3 inhibitors potently inhibited proliferation of the most abundant cell in the mammalian brain, the cerebellar granule cell progenitor (GCP). These studies identify a previously unappreciated role for GSK3ß mediated regulation of Wee1 during the cell cycle and in neurogenesis. Furthermore, they suggest that pharmacological inhibition of Wee1 may be therapeutically attractive in some cancers where GSK-3ß or Wee1 are dysregulated.


Subject(s)
Cell Cycle Proteins/metabolism , Cerebellum/cytology , Cytoplasmic Granules/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Animals , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/metabolism , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Luciferases/metabolism , Mice , Protein Kinase Inhibitors/chemistry , Protein Stability/drug effects
15.
Cell Cycle ; 14(8): 1274-81, 2015.
Article in English | MEDLINE | ID: mdl-25606665

ABSTRACT

Kinase signaling networks are well-established mediators of cell cycle transitions. However, how kinases interact with the ubiquitin proteasome system (UPS) to elicit protein turnover is not fully understood. We sought a means of identifying kinase-substrate interactions to better understand signaling pathways controlling protein degradation. Our prior studies used a luciferase fusion protein to uncover kinase networks controlling protein turnover. In this study, we utilized a similar approach to identify pathways controlling the cell cycle protein p27(Kip1). We generated a p27(Kip1)-luciferase fusion and expressed it in cells incubated with compounds from a library of pharmacologically active compounds. We then compared the relative effects of the compounds on p27(Kip1)-luciferase fusion stabilization. This was combined with in silico kinome profiling to identify potential kinases inhibited by each compound. This approach effectively uncovered known kinases regulating p27(Kip1) turnover. Collectively, our studies suggest that this parallel screening approach is robust and can be applied to fully understand kinase-ubiquitin pathway interactions.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Kinases/metabolism , Signal Transduction , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Humans , Leupeptins/pharmacology , Luciferases/genetics , Luciferases/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Ubiquitin/metabolism
16.
J Biol Chem ; 289(27): 18893-903, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24817118

ABSTRACT

Eukaryotic mitotic entry is controlled by Cdk1, which is activated by the Cdc25 phosphatase and inhibited by Wee1 tyrosine kinase, a target of the ubiquitin proteasome pathway. Here we use a reporter of Wee1 degradation, K328M-Wee1-luciferase, to screen a kinase-directed chemical library. Hit profiling identified CK1δ-dependent Wee1 degradation. Small-molecule CK1δ inhibitors specifically disrupted Wee1 destruction and arrested HeLa cell proliferation. Pharmacological inhibition, siRNA knockdown, or conditional deletion of CK1δ also reduced Wee1 turnover. Thus, these studies define a previously unappreciated role for CK1δ in controlling the cell cycle.


Subject(s)
Casein Kinase Idelta/metabolism , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteolysis , Amino Acid Sequence , Animals , Casein Kinase Idelta/antagonists & inhibitors , Cell Cycle/drug effects , Cell Cycle Proteins/chemistry , Drug Evaluation, Preclinical , HeLa Cells , Humans , Mice , Molecular Sequence Data , Nuclear Proteins/chemistry , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Protein-Tyrosine Kinases/chemistry , Proteolysis/drug effects , Small Molecule Libraries/pharmacology
17.
Epigenetics ; 9(4): 611-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24496381

ABSTRACT

Epigenetic proteins have recently emerged as novel anticancer targets. Among these, bromodomain and extra terminal domain (BET) proteins recognize lysine-acetylated histones, thereby regulating gene expression. Newly described small molecules that inhibit BET proteins BRD2, BRD3, and BRD4 reduce proliferation of NUT (nuclear protein in testis)-midline carcinoma, multiple myeloma, and leukemia cells in vitro and in vivo. These findings prompted us to determine whether BET proteins may be therapeutic targets in the most common primary adult brain tumor, glioblastoma (GBM). We performed NanoString analysis of GBM tumor samples and controls to identify novel therapeutic targets. Several cell proliferation assays of GBM cell lines and stem cells were used to analyze the efficacy of the drug I-BET151 relative to temozolomide (TMZ) or cell cycle inhibitors. Lastly, we performed xenograft experiments to determine the efficacy of I-BET151 in vivo. We demonstrate that BRD2 and BRD4 RNA are significantly overexpressed in GBM, suggesting that BET protein inhibition may be an effective means of reducing GBM cell proliferation. Disruption of BRD4 expression in glioblastoma cells reduced cell cycle progression. Similarly, treatment with the BET protein inhibitor I-BET151 reduced GBM cell proliferation in vitro and in vivo. I-BET151 treatment enriched cells at the G1/S cell cycle transition. Importantly, I-BET151 is as potent at inhibiting GBM cell proliferation as TMZ, the current chemotherapy treatment administered to GBM patients. Since I-BET151 inhibits GBM cell proliferation by arresting cell cycle progression, we propose that BET protein inhibition may be a viable therapeutic option for GBM patients suffering from TMZ resistant tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Brain Neoplasms/pathology , Case-Control Studies , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
18.
Epigenetics ; 8(8): 785-95, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23807265

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant adult brain tumor. Standard GBM treatment includes maximal safe surgical resection with combination radiotherapy and adjuvant temozolomide (TMZ) chemotherapy. Alarmingly, patient survival at five-years is below 10%. This is in part due to the invasive behavior of the tumor and the resulting inability to resect greater than 98% of some tumors. In fact, recurrence after such treatment may be inevitable, even in cases where gross total resection is achieved. The Cancer Genome Atlas (TCGA) research network performed whole genome sequencing of GBM tumors and found that GBM recurrence is linked to epigenetic mechanisms and pathways. Central to these pathways are epigenetic enzymes, which have recently emerged as possible new drug targets for multiple cancers, including GBM. Here we review GBM treatment, and provide a systems approach to identifying epigenetic drivers of GBM tumor progression based on temporal modeling of putative GBM cells of origin. We also discuss advances in defining epigenetic mechanisms controlling GBM initiation and recurrence and the drug discovery considerations associated with targeting epigenetic enzymes for GBM treatment.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Epigenesis, Genetic , Glioblastoma/genetics , Glioblastoma/therapy , Adult , Animals , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Drug Discovery , Gene Regulatory Networks , Glioblastoma/pathology , Humans , Mice , Molecular Targeted Therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Systems Biology , Temozolomide
19.
Orphanet J Rare Dis ; 6: 10, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21418619

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is one of the most devastating neurodegenerative diseases. Neurotrophic factors have been widely tested to counteract neurodegenerative conditions, despite their unspecific neuronal access. The non-toxic C-terminal fragment of the tetanus toxin (TTC) heavy chain has been studied not only as a carrier molecule to the CNS but also as a neuroprotective agent. Because the neurotrophic effects of BDNF have been demonstrated in vitro and in vivo, the question addressed in this work is whether a fusion molecule of BDNF-TTC may have a synergistic effect and enhance the neuroprotective properties of TTC alone in a mouse model of ALS. METHODS: Recombinant plasmid constructs (pCMV-TTC and pCMV-BDNF-TTC) were injected into the quadriceps femoris and triceps brachialis muscles of SOD1(G93A) transgenic mice at 8 weeks of age. The hanging wire and rotarod tests were performed to assess motor coordination, strength and balance. Electrophysiological tests, morphological assays of spinal cord sections of L2 and L4 segments, and gene and protein expression analyses were performed. The Kaplan-Meier survival analysis test was used for comparisons of survival. Multiple comparisons of data were analyzed using a one-way analysis of variance (ANOVA). RESULTS: Treatment with the fusion-molecule BDNF-TTC and with TTC alone significantly delayed the onset of symptoms and functional deficits of SOD1(G93A) mice. Muscle innervation was partially preserved with these treatments, and the number of surviving motoneurons in L2 spinal cord segment was increased particularly by the fusion protein induction. Inhibition of pro-apoptotic protein targets (caspase-3 and Bax) and significant phosphorylation of Akt and ERK were also found in the spinal cord of treated mice. CONCLUSIONS: Significant improvements in behavioral and electrophysiological results, motoneuron survival and anti-apoptotic/survival-activated pathways were observed with BDNF-TTC treatment. However, no synergistic effect was found for this fusion molecule. Although BDNF in the fusion molecule is capable of activating autocrine and neuroprotective pathways, TTC treatment alone yielded similar neuroprotection. Therefore, an accurate study of the neuroprotective effects of TTC fusion molecules should be performed to obtain a better understanding of its effects.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Brain-Derived Neurotrophic Factor/pharmacology , Genetic Therapy/methods , Recombinant Fusion Proteins/pharmacology , Tetanus Toxin/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Animals , Behavior, Animal/physiology , Female , Gene Expression Profiling , Histocytochemistry , Kaplan-Meier Estimate , Male , Mice , Mice, Transgenic , Motor Activity/physiology , Neural Conduction/physiology , Plasmids/genetics , Random Allocation , Spinal Cord/pathology
20.
J Neurotrauma ; 28(5): 831-40, 2011 May.
Article in English | MEDLINE | ID: mdl-21332255

ABSTRACT

Spinal root avulsion leads to a progressive loss of axotomized motoneurons (MNs). Nowadays, there is no effective treatment to prolong MN survival that could permit recovery as a result of delayed surgical repair. Administration of Sigma-1 receptor (Sig-1R) ligands has been reported to promote beneficial effects after several types of neural injury. In order to shed light of whether Sig-1R ligands could promote MN survival after root avulsion, L4-L5 spinal roots were unilaterally avulsed in adult rats and the Sig-1R agonist Pre084 was administered at different doses. The ventral spinal cords of the animals were studied from 3 to 21 days post-operation (DPO) by using histological, immunohistochemical, and Western blot techniques. Daily treatment with 0.25 mg/kg Pre084 significantly promoted MN survival (68% vs 43% in untreated rats) at 21 DPO, an effect that was antagonized by coadministration of BD1063, an antagonist of Sig-1R. There was a reduction in astroglial- associated immunoreactivity in rats treated with Pre084. Moreover, Pre084 produced an increase in the Sig-1R co-chaperone BiP within MNs, and an increase of GDNF expression by astrocytes in the ventral horn early after injury. Although the mechanisms promoting MN survival by Pre084 remain unclear, we hypothesize that it is mediated at least in part through the increase in these cytoprotective factors. Therefore, early application of Sig-1R agonist appears to be a promising therapy to improve MN survival after root avulsion.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Heat-Shock Proteins/biosynthesis , Morpholines/pharmacology , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Spinal Nerve Roots/injuries , Animals , Axotomy , Blotting, Western , Cell Survival/drug effects , Female , Immunohistochemistry , Motor Neurons/pathology , Radiculopathy/drug therapy , Radiculopathy/pathology , Rats , Rats, Sprague-Dawley , Receptors, sigma/agonists , Spinal Nerve Roots/drug effects , Sigma-1 Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...