Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Small ; 20(33): e2311507, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38856024

ABSTRACT

The immunosuppressive characteristics and acquired immune resistance can restrain the therapy-initiated anti-tumor immunity. In this work, an antibody free programmed death receptor ligand 1 (PD-L1) downregulator (designated as CeSe) is fabricated to boost photodynamic activated immunotherapy through cyclin-dependent kinase 5 (CDK5) inhibition. Among which, FDA approved photosensitizer of chlorin e6 (Ce6) and preclinical available CDK5 inhibitor of seliciclib (Se) are utilized to prepare the nanomedicine of CeSe through self-assembly technique without drug excipient. Nanoscale CeSe exhibits an increased stability and drug delivery efficiency, contributing to intracellular production of reactive oxygen species (ROS) for robust photodynamic therapy (PDT). The PDT of CeSe can not only suppress the primary tumor growth, but also induce the immunogenic cell death (ICD) to release tumor associated antigens. More importantly, the CDK5 inhibition by CeSe can downregulate PD-L1 to re-activate the systemic anti-tumor immunity by decreasing the tumor immune escape and therapy-induced acquired immune resistance. This work provides an antibody free strategy to activate systemic immune response for metastatic tumor treatment, which may accelerate the development of translational nanomedicine with sophisticated mechanism.


Subject(s)
B7-H1 Antigen , Cyclin-Dependent Kinase 5 , Immunotherapy , Photochemotherapy , Photochemotherapy/methods , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Immunotherapy/methods , Animals , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Humans , Cell Line, Tumor , Down-Regulation/drug effects , Mice , Neoplasms/therapy , Neoplasms/drug therapy , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Chlorophyllides
2.
Am J Physiol Cell Physiol ; 326(6): C1648-C1658, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38682237

ABSTRACT

The authors' previous research has shown the pivotal roles of cyclin-dependent kinase 5 (CDK5) and its regulatory protein p35 in nerve growth factor (NGF)-induced differentiation of sympathetic neurons in PC12 cells. During the process of differentiation, neurons are susceptible to environmental influences, including the effects of drugs. Metformin is commonly used in the treatment of diabetes and its associated symptoms, particularly in diabetic neuropathy, which is characterized by dysregulation of the sympathetic neurons. However, the impacts of metformin on sympathetic neuronal differentiation remain unknown. In this study, we investigated the impact of metformin on NGF-induced sympathetic neuronal differentiation using rat pheochromocytoma PC12 cells as a model. We examined the regulation of TrkA-p35/CDK5 signaling in NGF-induced PC12 differentiation. Our results demonstrate that metformin reduces NGF-induced PC12 differentiation by inactivating the TrkA receptor, subsequently inhibiting ERK and EGR1. Inhibition of this cascade ultimately leads to the downregulation of p35/CDK5 in PC12 cells. Furthermore, metformin inhibits the activation of the presynaptic protein Synapsin-I, a substrate of CDK5, in PC12 differentiation. In addition, metformin alters axonal and synaptic bouton formation by inhibiting p35 at both the axons and axon terminals in fully differentiated PC12 cells. In summary, our study elucidates that metformin inhibits sympathetic neuronal differentiation in PC12 cells by disrupting TrkA/ERK/EGR1 and p35/CDK5 signaling. This research contributes to uncovering a novel signaling mechanism in drug response during sympathetic neuronal differentiation, enhancing our understanding of the intricate molecular processes governing this critical aspect of neurodevelopment.NEW & NOTEWORTHY This study unveils a novel mechanism influenced by metformin during sympathetic neuronal differentiation. By elucidating its inhibitory effects from the nerve growth factor (NGF) receptor, TrkA, to the p35/CDK5 signaling pathways, we advance our understanding of metformin's mechanisms of action and emphasize its potential significance in the context of drug responses during sympathetic neuronal differentiation.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 5 , Metformin , Nerve Growth Factor , Neurons , Receptor, trkA , Animals , Metformin/pharmacology , Rats , PC12 Cells , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Receptor, trkA/metabolism , Receptor, trkA/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Cell Differentiation/drug effects , Signal Transduction/drug effects , Neurogenesis/drug effects , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Phosphotransferases
3.
Chemosphere ; 358: 142124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677614

ABSTRACT

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 5 , Metformin , Neocortex , Metformin/pharmacology , Animals , Female , Pregnancy , Neocortex/drug effects , Cyclin-Dependent Kinase 5/metabolism , Rats , Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Cell Differentiation/drug effects , Neurogenesis/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Signal Transduction/drug effects
4.
Cell Commun Signal ; 22(1): 233, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641599

ABSTRACT

BACKGROUND: Multiple neurodegenerative diseases are induced by the formation and deposition of protein aggregates. In particular, the microtubule-associated protein Tau leads to the development of so-called tauopathies characterized by the aggregation of hyperphosphorylated Tau within neurons. We recently showed that the constitutive activity of the serotonin receptor 7 (5-HT7R) is required for Tau hyperphosphorylation and aggregation through activation of the cyclin-dependent kinase 5 (CDK5). We also demonstrated physical interaction between 5-HT7R and CDK5 at the plasma membrane suggesting that the 5-HT7R/CDK5 complex is an integral part of the signaling network involved in Tau-mediated pathology. METHODS: Using biochemical, microscopic, molecular biological, computational and AI-based approaches, we investigated structural requirements for the formation of 5-HT7R/CDK5 complex. RESULTS: We demonstrated that 5-HT7R domains responsible for coupling to Gs proteins are not involved in receptor interaction with CDK5. We also created a structural model of the 5-HT7R/CDK5 complex and refined the interaction interface. The model predicted two conserved phenylalanine residues, F278 and F281, within the third intracellular loop of 5-HT7R to be potentially important for complex formation. While site-directed mutagenesis of these residues did not influence Gs protein-mediated receptor signaling, replacement of both phenylalanines by alanine residues significantly reduced 5-HT7R/CDK5 interaction and receptor-mediated CDK5 activation, leading to reduced Tau hyperphosphorylation and aggregation. Molecular dynamics simulations of 5-HT7R/CDK5 complex for wild-type and receptor mutants confirmed binding interface stability of the initial model. CONCLUSIONS: Our results provide a structural basis for the development of novel drugs targeting the 5-HT7R/CDK5 interaction interface for the selective treatment of Tau-related disorders, including frontotemporal dementia and Alzheimer's disease.


Subject(s)
Cyclin-Dependent Kinase 5 , Enzyme Activation , Receptors, Serotonin , Humans , Alzheimer Disease/metabolism , Cyclin-Dependent Kinase 5/chemistry , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Phosphorylation , Receptors, Serotonin/chemistry , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Signal Transduction
5.
Mol Neurobiol ; 60(5): 2426-2441, 2023 May.
Article in English | MEDLINE | ID: mdl-36656459

ABSTRACT

Alzheimer's disease (AD) pathogenesis feature progressive neurodegeneration, amyloid-ß plaque formation, and neurofibrillary tangles. Ample evidence has indicated the involvement of epigenetic pathways in AD pathogenesis. Here, we show that the expression of microRNA 650 (miR-650) is altered in brains from AD patients. Furthermore, we found that the processing of primary miR-650 to mature miR-650 is misregulated. Bioinformatic analysis predicted that miR-650 targets the expression of three AD-associated components: Apolipoprotein E (APOE), Presenilin 1 (PSEN1), and Cyclin-Dependent Kinase 5 (CDK5), and we have experimentally confirmed that miR-650 is able to significantly reduce the expression of APOE, PSEN1, and CDK5 in vitro. Importantly, the overexpression of miR-650 was further shown to significantly alter the CDK5 level and ameliorate AD pathologies in APP-PSEN1 transgenic mice. Overall, our results indicate that miR-650 influences AD pathogenesis through regulation of CDK5.


Subject(s)
Alzheimer Disease , MicroRNAs , Mice , Animals , Alzheimer Disease/pathology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Neurofibrillary Tangles/metabolism , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Apolipoproteins E/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Peptides/metabolism
6.
Ann Transl Med ; 10(16): 887, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36110999

ABSTRACT

Background: Tearing of inner layer of aorta causes aortic dissection (AD), a severe disease with high morbidity and mortality. The pathological development of AD partially results from apoptotic death of aortic endothelial cells (AECs), the trigger and the molecular regulation of which remain largely unknown. Cyclin-dependent kinase 5 (CDK5) was initially detected in the brain as a proline-directed serine/threonine protein kinase regulating neuronal cell cycle re-entry and arrest. The abnormal expression of CDK5 leads to apoptotic cell death following abortive cell cycle re-entry in some neuronal diseases. Although physiological and pathological roles of CDK5 have been widely investigated, the expression and function of CDK5 in AD have not been reported. Therefore, the aim of the present study was to discuss the expression and function of CDK5 in AD. Methods: Gene expression profiles were compared between AD tissues and normal aortic tissues using Gene Expression Omnibus (GEO) database with bioinformatic tools. Different cell types were isolated from the digested AD and normal aortic specimens by fluorescence-activated cell sorting (FACS). Gene expression in cells was quantified by quantitative reverse transcription polymerase chain reaction. Endothelial cells purified by FACS were transfected in vivo with plasmids. Cell growth was analyzed by Cell Counting Kit-8 assay. Cell apoptosis was analyzed by terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assay. Results: Analysis of gene profiles from AD tissues and normal aortic tissues using GEO database showed significant higher expression of CDK5 and its downstream regulated genes, proliferating cell nuclear antigen, cyclin B1, and B-cell lymphoma 2, which are regulators for cell cycle and apoptosis. Analysis of purified cells from AD and normal aortic specimens further confirmed this result and showed that the major source of CDK5 was endothelial cells. Depletion of CDK5 inhibited apoptosis of AECs, while the expression of CDK5 promoted apoptosis of AECs. Conclusions: CDK5 induces apoptosis of AECs to promote AD. CDK5 appears to be a promising novel target for preventing AD.

7.
Mol Brain ; 15(1): 53, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35701839

ABSTRACT

Dysfunction of striatal dopaminergic circuits has been implicated in motor impairment and Parkinson's disease (PD)-related circadian perturbations that may represent an early prodromal marker of PD. Cyclin-dependent kinase 5 (CDK5) negatively regulates dopamine signaling in the striatum, suggesting a critical role of CDK5 in circadian and sleep disorders. Here, we used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing to produce mice with a dorsal striatum (DS)-specific knockdown (KD) of the Cdk5 gene (referred to as DS-CDK5-KD mice) and investigate its role in vivo. DS-CDK5-KD mice exhibited deficits in locomotor activity and disturbances in activity/rest behavior. Additionally, Golgi staining of neurons in the DS revealed that CDK5 deletion reduced dendrite length and the number of functional synapses, which was confirmed by significant downregulation of MAP2, PSD-95, and synapsin I. Correlated with this, DS-CDK5-KD mice displayed reduced phosphorylation of Tau at Thr181. Furthermore, whole-cell patch-clamp recordings of green fluorescent protein-tagged neurons in the striatum of DS-CDK5-KD mice revealed a decreased frequency of spontaneous inhibitory postsynaptic currents and altered excitatory/inhibitory synaptic balance. Notably, anterograde labeling showed that CDK5 KD in the DS disrupted long-range projections to the secondary motor cortex, dorsal and ventral thalamic nuclei, and basolateral amygdala, which are involved in the regulation of motor and circadian rhythms in the brain. These findings support a critical role of CDK5 in the DS in maintaining the striatal neural circuitry underlying motor functions and activity/rest associated with circadian rhythms that are perturbed in neurodegenerative disorders.


Subject(s)
Circadian Rhythm , Cyclin-Dependent Kinase 5 , Animals , Corpus Striatum/metabolism , Cyclin-Dependent Kinase 5/metabolism , Dopamine , Down-Regulation , Mice , Synapses/metabolism
8.
Bull Exp Biol Med ; 172(6): 701-708, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35503584

ABSTRACT

It is known that the expression of the deubiquitinating enzyme BRCA1-BRCA2-containing complex subunit 3 (BRCC3) and cyclin-dependent protein kinase 5 (Cdk5) is increased in Parkinson's disease (both are involved in neuroinflammatory response). However, the regulatory mechanism of Cdk5 on the post-translational modification of BRCC3 remains unclear. Here we studied whether Cdk5 phosphorylates BRCC3. Phosphorylation of BRCC3 by Cdk5 was predicted by GPS 5.0 software. His-BRCC3 plasmid was constructed by cloning the BRCC3 gene into pGEX-6P-1 vector, and then His-BRCC3 fusion protein was induced with isopropyl ß-d-1-thiogalactopyranoside and purified using His-Tag affinity chromatography purification agarose. Phosphorylation of BRCC3 fusion protein by Cdk5 in vitro was detected by mass spectrometry and Western blotting. The results showed that multiple phosphorylation sites were predicted by GPS 5.0, and the His-BRCC3 fusion protein was successfully induced and purified. In vitro kinase assay, Western blotting, and mass spectrometry showed that Cdk5 can phosphorylate BRCC3. It has been demonstrated that protein kinase Cdk5 can phosphorylate the deubiquitinating enzyme BRCC3 in vitro, which provides new data for further study on the mechanism of neurodegeneration.


Subject(s)
Cyclin-Dependent Kinase 5 , Deubiquitinating Enzymes , Blotting, Western , Cyclin-Dependent Kinase 5/metabolism , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Humans , Parkinson Disease/metabolism , Phosphorylation
9.
Cancer ; 128(9): 1775-1786, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35143052

ABSTRACT

BACKGROUND: The objective of this study was to investigate the role and molecular mechanism of cyclin-dependent kinase 5 (CDK5) in regulating the growth of tongue squamous cell carcinoma (TSCC). METHODS: The authors used multiple methods to detect the levels of CDK5 expression in samples of TSCC and to explore the relation between CDK5 expression and various clinicopathologic factors. In vivo and in vitro cell experiments were performed to detect the proliferation, invasion, and migration of TSCC cells with CDK5 knockdown or overexpression. These studies verified that CDK5 regulates the occurrence and development of TSCC cells through the microRNA 513c-5p/cell division cycle 25B pathway. RESULTS: An elevated level of CDK5 expression in TSCC tissues was identified as an independent risk factor affecting TSCC growth and patient prognosis. Patients who had TSCC with low levels of CDK5 expression had a higher survival rate than those with high levels. Knockdown of CDK5 reduced the proliferation, migration, and invasion of TSCC cells both in vitro and in vivo. In addition, the authors observed that CDK5 regulated the growth of TSCC through the microRNA 513c-5p/cell division cycle C25B pathway. CONCLUSIONS: CDK5 functions as an oncogene in TSCC and might serve as a molecular marker for use in the diagnosis and treatment of TSCC. LAY SUMMARY: Tongue squamous cell carcinoma (TSCC) is 1 of the most common malignant tumors of the head and neck, and the survival rate of patients with tongue cancer has been very low. Therefore, it is important to study the molecular mechanism of TSCC progression to identify biomarkers that can be used to improve its clinical diagnosis and treatment. Cyclin-dependent kinase 5 (CDK5) is an atypical member of the cyclin-dependent kinase family and is involved in regulating the cell cycle. Changes in the cell cycle are of great significance for the occurrence and development of tumor cells; and, in recent years, increasing evidence has suggested that CDK5 exists in a disordered state in cancer cells. In this study, the authors demonstrate that CDK5 functions as an oncogene in TSCC and might serve as a molecular marker for use in the diagnosis and treatment of TSCC.


Subject(s)
Cyclin-Dependent Kinase 5 , MicroRNAs , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms , cdc25 Phosphatases , Cell Cycle , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tongue Neoplasms/genetics , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
10.
Eur J Neurosci ; 54(9): 7048-7062, 2021 11.
Article in English | MEDLINE | ID: mdl-34622493

ABSTRACT

Calcium influx into presynaptic terminals through voltage-gated Ca2+ channels triggers univesicular or multivesicular release of neurotransmitters depending on the characteristics of the release machinery. However, the mechanisms underlying multivesicular release (MVR) and its regulation remain unclear. Previous studies showed that in rat cerebellum, the cyclin-dependent kinase inhibitor roscovitine profoundly increases excitatory postsynaptic current (EPSC) amplitudes at granule cell (GC)-Purkinje cell (PC) synapses by enhancing the MVR of glutamate. This compound can also moderately augment the amplitude and prolong the decay time of inhibitory postsynaptic currents (IPSCs) at molecular layer interneuron (MLI)-PC synapses via MVR enhancement and GABA spillover, thus allowing for persistent activation of perisynaptic GABA receptors. The enhanced MVR may depend on the driving force for Cav 2.1 channel-mediated Ca2+ influx. To determine whether the distinct spatiotemporal dynamics of presynaptic Ca2+ influence MVR, we compared the effects of slow and fast Ca2+ chelators, that is, EGTA and BAPTA, respectively, on roscovitine-induced actions at GC-PC and MLI-PC synapses. Membrane-permeable EGTA-AM decreased GC-PC EPSC and MLI-PC IPSC amplitudes to a similar extent but suppressed the roscovitine-induced enhancement of EPSCs. In contrast, BAPTA-AM attenuated the effects of roscovitine on IPSCs. These results suggest that roscovitine augmented glutamate release by activating the release machinery located distally from the Cav 2.1 channel clusters, while it enhanced GABA release in a manner less dependent on those at distal sites. Therefore, the spatial relationships among Ca2+ channels, buffers, and sensors are critical determinants of the differential facilitatory actions of roscovitine on glutamatergic and GABAergic synapses in the cerebellar cortex.


Subject(s)
Cerebellum/drug effects , Roscovitine/pharmacology , Synapses , Synaptic Transmission , Animals , Calcium Channels, N-Type , Cerebellum/metabolism , Glutamic Acid , Neurotransmitter Agents , Presynaptic Terminals/drug effects , Rats
11.
Int J Mol Sci ; 22(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34360858

ABSTRACT

Tauopathies are neurodegenerative diseases characterized by abnormal metabolism of misfolded tau proteins and are progressive. Pathological phosphorylation of tau occurs in the retinal ganglion cells (RGCs) after optic nerve injuries. Cyclin-dependent kinase-5 (Cdk5) causes hyperphosphorylation of tau. To determine the roles played by Cdk5 in retinal degeneration, roscovitine, a Cdk5 inhibitor, was injected intravitreally after optic nerve crush (ONC). The neuroprotective effect of roscovitine was determined by the number of Tuj-1-stained RGCs on day 7. The change in the levels of phosphorylated tau, calpain-1, and cleaved α-fodrin was determined by immunoblots on day 3. The expression of P35/P25, a Cdk5 activator, in the RGCs was determined by immunohistochemistry. The results showed that roscovitine reduced the level of phosphorylated tau by 3.5- to 1.6-fold. Calpain-1 (2.1-fold) and cleaved α-fodrin (1.5-fold) were increased on day 3, suggesting that the calpain signaling pathway was activated. P35/P25 was accumulated in the RGCs that were poorly stained by Tuj-1. Calpain inhibition also reduced the increase in phosphorylated tau. The number of RGCs decreased from 2191 ± 178 (sham) to 1216 ± 122 cells/mm2 on day 7, and roscovitine preserved the level at 1622 ± 130 cells/mm2. We conclude that the calpain-mediated activation of Cdk5 is associated with the pathologic phosphorylation of tau.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Optic Nerve Injuries , Retinal Ganglion Cells , Tauopathies , tau Proteins/metabolism , Animals , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Phosphorylation , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Roscovitine/pharmacology , Tauopathies/metabolism , Tauopathies/pathology
12.
Ann Transl Med ; 9(10): 850, 2021 May.
Article in English | MEDLINE | ID: mdl-34164484

ABSTRACT

BACKGROUND: Peritonitis is a common complication in which the peritoneum becomes inflamed. Peroxisome proliferator-activated receptor (PPAR)γ agonists and extracellular signal-regulated kinases 1/2 (ERK1/2) inactivation have been found to restore damage caused by lipopolysaccharide-induced (LPS) inflammation. This study aimed to investigate the association between PPARγ and ERK1/2 in LPS-induced inflammation in peritonitis. METHODS: Human peritoneal mesothelial cells were maintained in Dulbecco's Modified Eagle Medium and treated with LPS under a series of different concentrations and treatment times. Cellular interleukins-1ßeta (IL-1ß), cellular interleukins-6 (IL-6), cellular interleukins-12 (IL-12) were measured by enzyme-linked immunosorbent assay (ELISA) assay. Expression or activation of cyclin-dependent kinase (CDK)5, ERK1/2, and PPARγ was detected using quantitative real-time PCR and/or western blot. RESULTS: LPS induced dose- and time-dependent increments in the cellular IL-1ß, IL-6, and IL-12 contents, cyclin-dependent kinase 5 (CDK5) expression, and PPARγSer273 phosphorylation. Treatment with 1 µg/mL LPS for 12 hours was the optimal experimental design for inflammation stimulation. The concentration of LPS over 1 µg/mL or treatment more than 12 hours reduced the inflammatory status. LPS stimulation also activated ERK1/2 and increased its interaction with CDK5. Further, ERK1/2 inhibition by AZD0364 prevented IL-1ß, IL-6, IL-12, and CDK5 expression, as well as activation of ERK1/2 and phosphorylation of PPARγ, induced by LPS. Knockdown of CDK5 using its siRNA caused similar changes as AZD0364, minus ERK1/2 inactivation. CONCLUSIONS: Our results suggested that LPS-induced inflammation in human peritoneal mesothelial cells can be partly suppressed by inhibiting the ERK1/2/CDK5/PPARγ axis.

13.
Life Sci ; 269: 119062, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33476635

ABSTRACT

AIMS: Cyclin-dependent kinase 5 (CDK5) is a potential target for the treatment of cerebral ischemia. CDK5 is one of the upstream regulators for Dynamin-related protein 1 (Drp1) phosphorylation. This study intends to discuss whether CDK5 inhibition conferring neuroprotection in cerebral ischemia through regulating Drp1 phosphorylation. MATERIALS AND METHODS: Mouse neuroblastoma N2a cells and N1E-115 cells were cultured and subjected to oxygen-glucose deprivation/reperfusion (OGDR). N2a cells and N1E-115 cells were treated with Roscovitine, a pharmacological inhibitor of CDK5, or transfected with CDK5 siRNA to knock down CDK5 expression. N2a cells were transfected with different plasmids (Drp1-Myc, the dephosphorylation-mimic mutant Drp1S616A-Myc and the phosphorylation-mimic mutant Drp1S616D-Myc). The expression of CDK5 and its activator p35, Drp1 and phosphorylated Drp1 on S616 was determined by western blot. The morphology of mitochondria was detected by immunofluorescence staining and the proportion of N2a cells with apoptosis was detected by flow cytometry analysis. KEY FINDINGS: Expression of CDK5, p35 and phosphorylated Drp1 on S616 was strongly upregulated after 4 h and 12 h reperfusion following 4 h oxygen-glucose deprivation (OGD) at protein level. CDK5 inhibition by pre-treated with Roscovitine or transfection with CDK5 siRNA significantly ameliorated OGDR induced mitochondrial fragmentation and apoptosis. Overexpression of the phosphorylation-mimic mutant Drp1S616D abrogated the protective effect of CDK5 inhibition against OGDR induced mitochondrial fragmentation and apoptosis. SIGNIFICANCE: Our data indicate that the neuroprotective effect of CDK5 inhibition against OGDR induced neuronal damage is Drp1S616 phosphorylation dependent. A better understanding of the neuroprotective mechanisms of CDK5 inhibition in cerebral ischemia will help to develop safe and efficacious drugs targeting CDK5 signaling for clinical use.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Dynamins/metabolism , Glucose/deficiency , Mitochondria/pathology , Neuroblastoma/prevention & control , Oxygen/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Dynamins/genetics , Mice , Mitochondria/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroprotective Agents , Phosphorylation , Reperfusion Injury/complications , Signal Transduction , Tumor Cells, Cultured
14.
Front Cell Neurosci ; 14: 235, 2020.
Article in English | MEDLINE | ID: mdl-32903692

ABSTRACT

Aberrant brain insulin signaling plays a critical role in the pathology of Alzheimer's disease (AD). Mitochondrial dysfunction plays a role in the progression of AD, with excessive mitochondrial fission in the hippocampus being one of the pathological mechanisms of AD. However, the molecular mechanisms underlying the progression of AD and mitochondrial fragmentation induced by aberrant brain insulin signaling in the hippocampal neurons are poorly understood. Therefore, we investigated the molecular mechanistic signaling associated with mitochondrial dynamics using streptozotocin (STZ), a diabetogenic compound, in the hippocampus cell line, HT-22 cells. In this metabolic dysfunctional cellular model, hallmarks of AD such as neuronal apoptosis, synaptic loss, and tau hyper-phosphorylation are induced by STZ. We found that in the mitochondrial fission protein Drp1, phosphorylation is increased in STZ-treated HT-22 cells. We also determined that inhibition of mitochondrial fragmentation suppresses STZ-induced AD-like pathology. Furthermore, we found that phosphorylation of Drp1 was induced by CDK5, and inhibition of CDK5 suppresses STZ-induced mitochondrial fragmentation and AD-like pathology. Therefore, these findings indicate that mitochondrial morphology and functional regulation may be a strategy of potential therapeutic for treating abnormal metabolic functions associated with the pathogenesis of AD.

15.
Neuroscience ; 438: 9-24, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32353462

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a regulator of axon growth and radial neuronal migration in the developing mouse brain, and it plays critical roles in cortical structure formation and brain function. However, the function of Cdk5 in cortico-cortical and cortico-sensorimotor networks in the adult remains largely unknown. In this study, we investigated the function of Cdk5 in the rostral secondary motor cortex (M2) in the male mouse using CRISPR/Cas9 gene editing and somatic brain transgenesis, to produce M2-specific knockdown of Cdk5 in neurons in the male mouse. Mouse deficient in Cdk5 in the M2 exhibited a reduction in both the number of functional synapses and the total basal dendritic length, as well as motor dysfunction. Furthermore, whole-cell patch-clamp recordings in layer V green fluorescent protein (GFP)-tag pyramidal neurons revealed a decrease in the frequency and amplitude of miniature EPSCs and miniature IPSCs, as well as a reduction in the population synaptic responses (fEPSPs) in these mice. Specifically, retrograde labeling showed that Cdk5 knockdown in the M2 caused a reduction in long-range projections to the M2 from the thalamus/prefrontal cortex and claustrum. Collectively, our findings show a new regulatory role of Cdk5 in neural circuit maintenance, and that the changes in neural transmission and circuits in the mice with Cdk5 knockdown in the M2 likely contribute to the motor dysfunction in these animals.


Subject(s)
Cyclin-Dependent Kinase 5 , Motor Cortex , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Male , Mice , Motor Cortex/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Synaptic Transmission
16.
Eur J Neurosci ; 52(3): 3002-3021, 2020 08.
Article in English | MEDLINE | ID: mdl-32383214

ABSTRACT

Synaptic vesicle exocytosis is triggered by Ca2+ influx through several subtypes of voltage-gated calcium channels in the presynaptic terminal. We previously reported that paired-pulse stimulation at brief intervals increases Cav 2.1 (P/Q-type) channel-mediated multivesicular release (MVR) at glutamatergic synapses between granule cells (GCs) and molecular layer interneurons (MLIs) in rat cerebellar slices. However, it has yet to be determined how Cav 2 channel subtypes take part in MVR in single axon terminal. This study therefore aimed at examining the effects of roscovitine on different types of cerebellar synapses that make contacts with Purkinje cells (PCs), because this compound has been shown to enhance Cav 2.1 channel-mediated MVR at GC-MLI synapses. Bath application of roscovitine profoundly increased the amplitude of excitatory postsynaptic currents (EPSCs) at GC-PC synapses by a presynaptic mechanism as previously observed at GC-MLI synapses, whereas it caused a marginal effect on climbing fiber-mediated EPSCs in PCs. At MLI-PC synapses, roscovitine increased both the amplitude and decay time of inhibitory postsynaptic currents (IPSCs) by enhancing multivesicular GABA release. When extracellular Ca2+ concentration ([Ca2+ ]e ) decreased, roscovitine became less effective in increasing GC-PC EPSCs. By contrast, roscovitine was able to augment MLI-PC IPSCs in the low [Ca2+ ]e . The Cav 2.1 channel blocker ω-agatoxin IVA suppressed the roscovitine-induced facilitatory actions on both GC-PC EPSCs and MLI-PC IPSCs. These results demonstrate that roscovitine enhances MVR at the GC-PC excitatory synapses in a manner dependent on the driving force of Cav 2.1 channel-mediated Ca2+ influx into the nerve terminal, while it also facilitates MLI-PC inhibitory transmission via Ca2+ -insensitive mechanisms.


Subject(s)
Purkinje Cells , Synaptic Transmission , Animals , Cerebellum , Rats , Roscovitine , Synapses
17.
Acta Pharm Sin B ; 10(2): 358-373, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32082979

ABSTRACT

Blocking the programmed death-ligand 1 (PD-L1) on tumor cells with monoclonal antibody therapy has emerged as powerful weapon in cancer immunotherapy. However, only a minority of patients presented immune responses in clinical trials. To develop an alternative treatment method based on immune checkpoint blockade, we designed a novel and efficient CRISPR-Cas9 genome editing system delivered by cationic copolymer aPBAE to downregulate PD-L1 expression on tumor cells via specifically knocking out Cyclin-dependent kinase 5 (Cdk5) gene in vivo. The expression of PD-L1 on tumor cells was significantly attenuated by knocking out Cdk5, leading to effective tumor growth inhibition in murine melanoma and lung metastasis suppression in triple-negative breast cancer. Importantly, we demonstrated that aPBAE/Cas9-Cdk5 treatment elicited strong T cell-mediated immune responses in tumor microenvironment that the population of CD8+ T cells was significantly increased while regulatory T cells (Tregs) was decreased. It may be the first case to exhibit direct in vivo PD-L1 downregulation via CRISPR-Cas9 genome editing technology for cancer therapy. It will provide promising strategy for preclinical antitumor treatment through the combination of nanotechnology and genome engineering.

18.
Curr Top Med Chem ; 20(1): 4-36, 2020.
Article in English | MEDLINE | ID: mdl-31797761

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disease that 4 widespread in the elderly. The etiology of AD is complicated, and its pathogenesis is still unclear. Although there are many researches on anti-AD drugs, they are limited to reverse relief symptoms and cannot treat diseases. Therefore, the development of high-efficiency anti-AD drugs with no side effects has become an urgent need. Based on the published literature, this paper summarizes the main targets of AD and their drugs, and focuses on the research and development progress of these drugs in recent years.


Subject(s)
Alzheimer Disease/drug therapy , Drug Delivery Systems , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Humans
19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-815605

ABSTRACT

@# Objective: To study the expression of miR-142-5p in lung adenocarcinoma tissues, and to explore its effect on proliferation, invasion, migration and epithelieal-mesenchymal transition (EMT) of H1650 cells and the potential mechanisms. Methods:Atotal of 107 pairs of lung adenocarcinoma tissues and corresponding para-cancerous tissues from patients, who underwent tumor resection and were pathologically confirmed at the Department of Thoracic Surgery, the Fourth Hospital of Hebei Medical University between Jan. 2014 and Jan. 2015, were collected for this study; in addition, human lung adenocarcinoma cell lines (H1650, HCC827, A549, H1975, PC9) and human bronchial epithelial BEAS-2B cells were also used in this study. qPCR was used to detect the expression of miR-142-5p in lung adenocarcinoma tissues and cell lines. The correlation between expression of miR-142-5p and clinical features was analyzed.After transfection with miR-142-5p mimics or miR-negative control (miR-NC) plasmid, the proliferation, invasion and migration of H1650 cells were detected with CCK-8, Transwell invasion assay and Wound healing assay, respectively. The bioinforamtics tool was used to predict the target genes of miR-142-5p, and Luciferase reporter gene assay was performed to validate the regulation of miR-142-5p on target gene. Western blotting (WB) was used to detect the expressions of cyclin-dependent kinase 5 (CDK5) and EMTrelated protein. Results: Compared to Para-cancerous tissues and BEAS-2B cells, the expression of miR-142-5p was lower in lung adenocarcinoma tissues and cell lines (all P<0.01). Of the 107 cases of lung adenocarcinoma tissues, 61 cases (57.01%) showed decreased miR-142-5 expression, which was correlated with the TNM stage and lymph node metastasis (both P<0.01). Transfection of miR-142-5p mimics significantly up-regulated the expression of miR-142-5p and decreased the proliferation, invasion and migration of H1650 cells (all P<0.05 or P<0.01). Bioinformatics showed that CDK5 was a target gene of miR-142-5p. Luciferase reporter gene assay and WB validated that miR-142-5p could significantly down-regulate CDK5 expression in H1650 cells, up-regulate the expression of E-cadherin and down-regulate the expressions of N-cadherin, Twist and Snail in H1650 cells (all P<0.01). Conclusion: miR-142-5p is low expressed in lung adenocarcinoma tissues and cell lines; it suppresses the EMT process to inhibit, invasion and migration of H1650 cells via down-regulating the expression of CDK5.

20.
Acta Pharmaceutica Sinica B ; (6): 358-373, 2020.
Article in English | WPRIM (Western Pacific) | ID: wpr-787623

ABSTRACT

Blocking the programmed death-ligand 1 (PD-L1) on tumor cells with monoclonal antibody therapy has emerged as powerful weapon in cancer immunotherapy. However, only a minority of patients presented immune responses in clinical trials. To develop an alternative treatment method based on immune checkpoint blockade, we designed a novel and efficient CRISPR-Cas9 genome editing system delivered by cationic copolymer aPBAE to downregulate PD-L1 expression on tumor cells specifically knocking out Cyclin-dependent kinase 5 () gene . The expression of PD-L1 on tumor cells was significantly attenuated by knocking out , leading to effective tumor growth inhibition in murine melanoma and lung metastasis suppression in triple-negative breast cancer. Importantly, we demonstrated that aPBAE/Cas9-Cdk5 treatment elicited strong T cell-mediated immune responses in tumor microenvironment that the population of CD8 T cells was significantly increased while regulatory T cells (Tregs) was decreased. It may be the first case to exhibit direct PD-L1 downregulation CRISPR-Cas9 genome editing technology for cancer therapy. It will provide promising strategy for preclinical antitumor treatment through the combination of nanotechnology and genome engineering.

SELECTION OF CITATIONS
SEARCH DETAIL