Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 255
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884454

ABSTRACT

Cord blood T cells (CBTC) from a proportion of newborns express low/deficient levels of some protein kinase C (PKC) isozymes, with low levels of PKCζ correlating with increased risk of developing allergy and associated decrease in interferon-gamma (IFN-γ) producing T cells. Interestingly, these lower levels of PKCζ were increased/normalized by supplementing women during pregnancy with n-3 polyunsaturated fatty acids. However, at present, we have little understanding of the transient nature of the deficiency in the neonate and how PKCζ relates to other PKC isozymes and whether their levels influence maturation into IFN-γ producing T cells. There is also no information on PKCζ isozyme levels in the T cell subpopulations, CD4+ and CD8+ cells. These issues were addressed in the present study using a classical culture model of neonatal T cell maturation, initiated with phytohaemagglutinin (PHA) and recombinant human interleukin-2 (rhIL-2). Of the isozymes evaluated, PKCζ, ß2, δ, µ, ε, θ and λ/ι were low in CBTCs. The PKC isozyme deficiencies were also found in the CD4+ and CD8+ T cell subset levels of the PKC isozymes correlated between the two subpopulations. Examination of changes in the PKC isozymes in these deficient cells following addition of maturation signals showed a significant increase in expression within the first few hours for PKCζ, ß2 and µ, and 1-2 days for PKCδ, ε, θ and λ/ι. Only CBTC PKCζ isozyme levels correlated with cytokine production, with a positive correlation with IFN-γ, interleukin (IL)-2 and tumour necrosis factor-alpha (TNF), and a negative association with IL-9 and IL-10. The findings reinforce the specificity in using CBTC PKCζ levels as a biomarker for risk of allergy development and identify a period in which this can be potentially 'corrected' after birth.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Fetal Blood/cytology , Protein Kinase C/genetics , Female , Fetal Blood/immunology , Gestational Age , Humans , Infant, Newborn , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-9/metabolism , Male , Phytohemagglutinins/pharmacology , Pregnancy , Tumor Necrosis Factor-alpha/metabolism
2.
Nat Commun ; 12(1): 2507, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947863

ABSTRACT

Notch1 is a crucial oncogenic driver in T-cell acute lymphoblastic leukemia (T-ALL), making it an attractive therapeutic target. However, the success of targeted therapy using γ-secretase inhibitors (GSIs), small molecules blocking Notch cleavage and subsequent activation, has been limited due to development of resistance, thus restricting its clinical efficacy. Here, we systematically compare GSI resistant and sensitive cell states by quantitative mass spectrometry-based phosphoproteomics, using complementary models of resistance, including T-ALL patient-derived xenografts (PDX) models. Our datasets reveal common mechanisms of GSI resistance, including a distinct kinase signature that involves protein kinase C delta. We demonstrate that the PKC inhibitor sotrastaurin enhances the anti-leukemic activity of GSI in PDX models and completely abrogates the development of acquired GSI resistance in vitro. Overall, we highlight the potential of proteomics to dissect alterations in cellular signaling and identify druggable pathways in cancer.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Oligopeptides/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase C/metabolism , Receptor, Notch1/antagonists & inhibitors , Acetophenones/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzopyrans/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Drug Resistance, Neoplasm/genetics , Gene Ontology , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Phosphorylation , Protein Array Analysis , Protein Biosynthesis/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinases/metabolism , Proteomics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry , Xenograft Model Antitumor Assays
3.
Aging (Albany NY) ; 13(9): 12466-12478, 2021 04 25.
Article in English | MEDLINE | ID: mdl-33896796

ABSTRACT

Activated protein C (APC) is an anticoagulant with potent cytoprotective and anti-inflammatory effects. K150del, a natural variant of APC, is associated with reduced anticoagulant activity. We performed a comprehensive study to analyze the functional alterations of the K150del mutant. Transcriptome analysis of HEK 293T cells treated with wild and mutant APC revealed differentially expressed genes enriched in inflammatory, apoptotic, and virus defense-related signaling pathways. Both wild and mutant APC displayed concentration-dependent cytoprotective effects. Low concentrations of K150del mutant resulted in decreased anti-inflammatory and anti-apoptotic activities, whereas its higher concentrations restored these effects. Expression of virus defense-related genes improved in mouse lung tissues after repeated administration of the APC variant. These results suggest that the APC K150del mutant could help clinicians to accurately predict disease risks and serve as a potential auxiliary therapeutic in viral infections, including 2019 coronavirus disease (COVID-19).


Subject(s)
COVID-19 , Protein Kinase C/genetics , Protein Kinase C/metabolism , Animals , HEK293 Cells , Humans , Mice , Polymorphism, Single Nucleotide , SARS-CoV-2
4.
Zhongguo Zhong Yao Za Zhi ; 46(3): 599-604, 2021 Feb.
Article in Chinese | MEDLINE | ID: mdl-33645025

ABSTRACT

Protein kinase C(PKC) is a type of protein kinase widely involved in cell proliferation and development, but the developmental mechanism in the gonads of androgynous animals is still unclear. In order to explore the role of protein kinase C in the development of Whitmania pigra germ cells, the Wh. pigra PKC(Wp-PKC) gene was cloned, bioinformatics analysis was conducted, and fluorescent quantitative PCR was used to analyze the expression of female and male gonads. The results showed that:(1)The cloned Wp-PKC had a full length of 2 580 bp, a relative molecular weight of 76 555.19, and contains an open reading frame encoding 670 amino acids, Wp-PKC was closely related to Danio rerio PKC-α and rat PKC-γ. The similarity of amino acid sequence was 55% and 58%.(2)The protein encoded by Wp-PKC had no signal peptide and was a hydrophilic protein. The secondary structure is mainly composed of random coils, α-helices, extended chains, folds and folds, with the largest proportion of random coils and α-helices. Wp-PKC protein does not contain a transmembrane domain. Multiple sequence alignment and domain prediction analysis show that Wp-PKC contains 4 conserved domains of classical protein kinase C.(3)Fluorescence quantitative results showed that the expression of Wp-PKC in Wh. pigra gonads was positively correlated with the development of germ cells, and the expression in male gonads was significantly higher than that in female gonads. In summary, Wp-PKC is a classic PKC, and Wp-PKC may promote the development of Wh. pigra, especially the development of male gonads, and provide references for further research on the developmental mechanisms of Wh. pigra.


Subject(s)
Leeches , Animals , Cloning, Molecular , Female , Gonads , Leeches/genetics , Male , Ovary , Protein Kinase C/genetics , Rats
5.
J Ethnopharmacol ; 268: 113654, 2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33271248

ABSTRACT

HEADINGS ETHNOPHARMACOLOGICAL RELEVANCE: Sophora alopecuroides L. is a traditional ethnopharmacological plant, which is widely used in traditional Chinese medicine and Mongolian and Uighur medicine to ameliorate "thirst disease". AIM OF THE STUDY: This study aimed to investigate the antidiabetic activities and mechanisms of a flavonoid-rich extract from Sophora alopecuroides L. (SA-FRE) both in vivo and vitro. MATERIALS AND METHODS: The main six chemical constituents of SA-FRE were elucidated based on an off-line semi-preparative liquid chromatography nuclear magnetic resonance (LC-NMR) protocol. Myc-GLUT4-mOrange-L6 cell models and mouse model with diabetes induced by high-fat diet combined with STZ injection were respectively adopted to investigate the antidiabetic effects of SA-FRE both in vitro and vivo. RESULTS: In vivo, 4-week treatment of SA-FRE ameliorated hyperglycemia, dyslipidemia, and insulin resistance in diabetic mice. Mechanically, SA-FRE regulated PPARα and PPARγ expression in white adipose tissue (WAT) and liver, thereby ameliorating dyslipidemia. Moreover, SA-FRE increased the phosphorylation of PKC and further stimulated the GLUT4 expression in WAT and skeletal muscle, thus increasing the glucose utilization in vivo. In vitro, 50 µg/mL SA-FRE increased GLUT4 translocation to about 1.91-fold and glucose uptake to 1.82-fold in L6-myotubes. SA-FRE treatment increased the GLUT4 expression at both gene and protein levels. Furthermore, only Gö6983, a PKC inhibitor, reversed the SA-FRE-induced GLUT4 translocation and expression at the gene and protein levels. CONCLUSIONS: Generally, SA-FRE ameliorated hyperglycemia, dyslipidemia, and insulin resistance partly through activating PKC/GLUT4 pathway and regulating PPARα and PPARγ expression.


Subject(s)
Glucose Transporter Type 4/biosynthesis , Hypoglycemic Agents/therapeutic use , PPAR alpha/biosynthesis , PPAR gamma/biosynthesis , Protein Kinase C/biosynthesis , Sophora , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat/adverse effects , Flavonoids/isolation & purification , Flavonoids/pharmacology , Flavonoids/therapeutic use , Gene Expression , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , PPAR alpha/genetics , PPAR gamma/genetics , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Protein Kinase C/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Streptozocin
6.
Cell Signal ; 77: 109819, 2021 01.
Article in English | MEDLINE | ID: mdl-33147518

ABSTRACT

Current treatment options of glioblastoma include chemotherapy and limited surgical resection. Temozolomide (TMZ) is the current therapeutic choice for chemotherapy. Still, it has severe limitations due to the development of resistance that occurs by genetic modification and constitutive activation of several cell signaling pathways. Therefore, it is essential to develop combination therapy of TMZ with other novel compounds to prevent the development of chemo-resistance. In this study, we used two inhibitors; ICA, an inhibitor of PKC-ι and ζ-Stat, an inhibitor of PKC-ζ. T98G and U87MG glioblastoma cells were treated with either ICA or ζ-stat or TMZ monotherapies, as well as TMZ were combined with either ICA or ζ-stat for five consecutive days. Our in vitro results exhibited that ICA when combined with TMZ, significantly decreased the viability of cancerous cells compared with untreated or TMZ or ICA monotherapies. Additionally, glioblastoma cells were remarkably undergoing apoptosis against the combination treatment of TMZ and ICA nucleotide compared with untreated control cells, as suggested by our Annexin-V/PI flow cytometric analysis. Moreover, the combination of TMZ and ICA also decreased the invasion of glioblastoma cell lines by acting on FAK/Paxillin pathway, as evidenced by scratch assay, transwell invasion assay, Western blot and immunoprecipitation analysis. Furthermore, our in vivo data presented that the combination of ICA and TMZ also reduced glioblastoma tumor growth and volume in mice. These data suggest that atypical PKCs, particularly PKC-ι might be an important therapeutic target as adjuvant therapy in the treatment of glioblastoma.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Temozolomide/pharmacology , Actin Cytoskeleton/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Therapy, Combination , Focal Adhesion Kinase 1/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Mice , Mice, Nude , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Temozolomide/therapeutic use , Transplantation, Heterologous
7.
Food Funct ; 11(11): 9858-9867, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33089839

ABSTRACT

3ß,7ß,25-Trihydroxycucurbita-5,23(E)-dien-19-al (TCD) is a triterpenoid isolated from wild bitter gourd that is a common tropical vegetable with neuroprotective effects. Because excessive glutamate release is a major cause of neuronal damage in various neurological disorders, the aims of this study were to examine the effect of TCD on glutamate release in vitro and to examine the effect of TCD in vivo. In rat cerebrocortical synaptosomes, TCD reduced 4-aminopyridine (4-AP)-stimulated glutamate release and Ca2+ concentration elevation, but had no effect on plasma membrane potential. TCD-mediated inhibition of 4-AP-induced glutamate release was dependent on the presence of extracellular calcium; persisted in the presence of the glutamate transporter inhibitor dl-TBOA, P/Q-type Ca2+ channel blocker ω-agatoxin IVA, and intracellular Ca2+-releasing inhibitors dantrolene and CGP37157; and was blocked by the vesicular transporter inhibitor bafilomycin A1 and the N-type Ca2+ channel blocker ω-conotoxin GVIA. Molecular docking studies have demonstrated that TCD binds to N-type Ca2+ channels. TCD-mediated inhibition of 4-AP-induced glutamate release was abolished by the Ca2+-dependent protein kinase C (PKC) inhibitor Go6976, but was unaffected by the Ca2+-independent PKC inhibitor rottlerin. Furthermore, TCD considerably reduced the phosphorylation of PKC, PKCα, and myristoylated alanine-rich C kinase substrate, a major presynaptic substrate for PKC. In a rat model of kainic acid (KA)-induced excitotoxicity, TCD pretreatment substantially attenuated KA-induced neuronal death in the CA3 hippocampal region. These results suggest that TCD inhibits synaptosomal glutamate release by suppressing N-type Ca2+ channels and PKC activity and exerts protective effects against KA-induced excitotoxicity in vivo.


Subject(s)
Glutamic Acid/metabolism , Kainic Acid/adverse effects , Momordica charantia/chemistry , Nervous System Diseases/drug therapy , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Synaptosomes/drug effects , Triterpenes/administration & dosage , Animals , Calcium/metabolism , Calcium Channel Blockers/administration & dosage , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Death/drug effects , Humans , Male , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism
8.
Mol Pain ; 16: 1744806920943685, 2020.
Article in English | MEDLINE | ID: mdl-32865105

ABSTRACT

Neuropathic pain can be generated by chronic compression of dorsal root ganglion (CCD). Stimulation of primary motor cortex can disrupt the nociceptive sensory signal at dorsal root ganglion level and reduce pain behaviors. But the mechanism behind it is still implicit. Protein kinase C gamma is known as an essential enzyme for the development of neuropathic pain, and specific inhibitor of protein kinase C gamma can disrupt the sensory signal and reduce pain behaviors. Optogenetic stimulation has been emerged as a new and promising conducive method for refractory neuropathic pain. The aim of this study was to provide evidence whether optical stimulation of primary motor cortex can modulate chronic neuropathic pain in CCD rat model. Animals were randomly divided into CCD group, sham group, and control group. Dorsal root ganglion-compressed neuropathic pain model was established in animals, and knocking down of protein kinase C gamma was also accomplished. Pain behavioral scores were significantly improved in the short hairpin Protein Kinase C gamma knockdown CCD animals during optic stimulation. Ventral posterolateral thalamic firing inhibition was also observed during light stimulation on motor cortex in CCD animal. We assessed alteration of pain behaviors in pre-light off, stimulation-light on, and post-light off state. In vivo extracellular recording of the ventral posterolateral thalamus, viral expression in the primary motor cortex, and protein kinase C gamma expression in dorsal root ganglion were investigated. So, optical cortico-thalamic inhibition by motor cortex stimulation can improve neuropathic pain behaviors in CCD animal, and knocking down of protein kinase C gamma plays a conducive role in the process. This study provides feasibility for in vivo optogenetic stimulation on primary motor cortex of dorsal root ganglion-initiated neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Motor Cortex/metabolism , Neuralgia/metabolism , Optogenetics/methods , Protein Kinase C/metabolism , Thalamus/metabolism , Animals , Behavior Rating Scale , Behavior, Animal/physiology , Female , Ganglia, Spinal/enzymology , Ganglia, Spinal/injuries , Gene Knockdown Techniques , Immunohistochemistry , Motor Cortex/enzymology , Motor Cortex/radiation effects , Neuralgia/genetics , Optical Fibers , Protein Kinase C/genetics , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Thalamus/enzymology
9.
Mol Cell Endocrinol ; 518: 110991, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32841709

ABSTRACT

The function of the gonadotropin-releasing hormone (GnRH) neuron is critical to maintain reproductive function and a significant decrease in GnRH can lead to disorders affecting fertility, including hypogonadotropic hypogonadism. Spexin (SPX) is a novel hypothalamic neuropeptide that exerts inhibitory effects on reproduction and feeding by acting through galanin receptor 2 (GALR2) and galanin receptor 3 (GALR3). Fatty acids can act as nutritional signals that regulate the hypothalamic-pituitary-gonadal (HPG) axis, and elevated levels of circulating saturated fatty acids associated with high fat diet (HFD)-feeding have been shown to induce neuroinflammation, endoplasmic reticulum stress and hormonal resistance in the hypothalamus, as well as alter neuropeptide expression. We previously demonstrated that palmitate, the most common saturated fatty acid in a HFD, elevates the expression of Spx, Galr2 and Galr3 mRNA in a model of appetite-regulating neuropeptide Y hypothalamic neurons. Here, we found that Spx, Galr2 and Galr3 mRNA were also significantly induced by palmitate in a model of reproductive GnRH neurons, mHypoA-GnRH/GFP. As a follow-up to our previous report, we examined the molecular pathways by which Spx and galanin receptor mRNA was regulated in this cell line. Furthermore, we performed inhibitor studies, which revealed that the effect of palmitate on Spx and Galr3 mRNA involved activation of the innate immune receptor TLR4, and we detected differential regulation of the three genes by the protein kinases PKC, JNK, ERK, and p38. However, the intracellular metabolism of palmitate to ceramide did not appear to be involved in the palmitate-mediated gene regulation. Overall, this suggests that SPX may play a role in reproduction at the level of the hypothalamus and the pathways by which Spx, Galr2 and Galr3 are altered by fatty acids could provide insight into the mechanisms underlying reproductive dysfunction in obesity.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/cytology , Palmitates/pharmacology , Peptide Hormones/genetics , Receptor, Galanin, Type 2/genetics , Receptor, Galanin, Type 3/genetics , Animals , Cell Line , Female , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Peptide Hormones/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptor, Galanin, Type 2/metabolism , Receptor, Galanin, Type 3/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Up-Regulation
10.
J Agric Food Chem ; 68(31): 8185-8194, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32692177

ABSTRACT

Blueberry anthocyanin-rich extract (BAE) was supplemented to high-fat diet (HFD)-fed mice to investigate sphingolipid metabolism modulating factors involved in the attenuated hyperinsulinemia and hyperlipidemia. A BAE-containing diet effectively controlled food intake and liver weight and significantly attenuated insulin resistance triggered by a HFD. Higher BAE (200 mg/kg of body weight) administration performed more efficiently in the improvement of hepatic steatosis and adipocyte hypertrophy, together with distinct suppressions in serum triacylglycerol and cholesterol in total and species. Serum lipid compositions revealed 200 mg/kg of BAE supplementation remarkably suppressed ceramide accumulation. Consistently, genes encoding enzymes associated with sphingomyelin conversion and ceramide de novo synthesis were modulated toward a healthy direction for restrained sphingolipid accumulation. Further, the inhibited mRNA expressions of protein phosphatase 2A and protein kinase Cζ involved in blocking Akt phosphorylation connected the controlled ceramides with the restored insulin sensitivity.


Subject(s)
Anthocyanins/administration & dosage , Blueberry Plants/chemistry , Ceramides/blood , Hyperlipidemias/drug therapy , Insulin Resistance , Plant Extracts/administration & dosage , Animals , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Fruit/chemistry , Humans , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/genetics , Male , Mice , Mice, Inbred C57BL , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Triglycerides/metabolism
11.
Curr Med Sci ; 40(2): 354-362, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32337697

ABSTRACT

Imatinib mesylate (IM) is the first-line treatment for Philadelphia (Ph) chromosomal positive leukemia by inhibiting phosphorylation of substrates via binding to the ABL kinase domain. Because of the drug resistance, side effects and the high cost of IM, it is necessary to find anti-cancer drugs with relatively low toxicity and cost, and enhanced efficacy, such as traditional Chinese medicines (TCMs). As one of TCMs, Huai Qi Huang (HQH) was chosen to treat BV173 and K562 cells. Various concentrations of HQH were added to cells for 24-72 h. Co-treatment of HQH and trametinib, an MEK inhibitor, was used to verify the synergistic effects on cell viability and apoptosis. Knockdown and overexpression of mitogen-activated protein kinase kinase 4 (MEK4) were implemented to demonstrate the role of MEK in cell apoptosis. Cell viability and apoptosis were measured by cell counting kit-8 assay (CCK8) and flow cytometry, respectively. Western blotting and real-time quantitative PCR (RT-qPCR) were used to assess protein and mRNA expression levels, respectively. The results showed that HQH inhibited survival and promoted apoptosis of BV173 and K562 cells in a dose-dependent manner, accompanied with down-regulation of PRKCH mRNA as well as CRAF, MEK4, phospho-ERK (pERK) and BCL2 proteins, and up-regulation of cleaved caspase3 protein. Co-treatment of HQH and trametinib had a synergistic effect on inhibiting survival and promoting apoptosis. MEK4 knockdown increased apoptosis, and had a synergistic effect with HQH. In contrast, MEK4 overexpression decreased apoptosis, and had the opposite effect with HQH. Collectively, the results of this study may identify a therapeutic mechanism of HQH on promoting apoptosis, and provide a potential option for treatment of Ph+ leukemia.


Subject(s)
Down-Regulation , Drugs, Chinese Herbal/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Protein Kinase C/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MAP Kinase Signaling System/drug effects , Protein Kinase C/genetics
12.
Curr Mol Med ; 19(8): 570-578, 2019.
Article in English | MEDLINE | ID: mdl-31210105

ABSTRACT

BACKGROUND: This study was conducted to determine the effects of vitamin D supplementation on some of the gene expressions related to insulin and lipid metabolism in diabetic hemodialysis (HD) patients. METHODS: A double-blind, randomized, placebo-controlled clinical trial was carried out in 55 patients with diabetic HD. The current project used two groups in which each subject received vitamin D supplements (50,000 IU, n=28) or placebo (50,000 IU, n=27) every 2 weeks for 12 weeks. Gene expression analyses (RT-PCR) were included to obtain the rate of gene expression of the related insulin and lipid metabolism genes in peripheral blood mononuclear cells (PBMCs) of patients with diabetic HD. RESULTS: Our data revealed that consumption of vitamin D supplementation enables to overexpress the peroxisome proliferation-activated receptor gamma (PPAR-γ) (P=0.001), AKT (P=0.04), PI3K (P=0.02), insulin receptor substrate-1 (IRS1) (P0.008) and glucose transporter type 4 (GLUT-4) (P=0.01) and downregulate the expression of protein kinase C (PKC) (P=0.001) in patients with diabetic HD than control group following the 12-week intervention. In addition, vitamin D supplementation downregulated low-density lipoprotein receptor (LDLR) (P=0.03) expression in the subjects with diabetic HD than the control group. Vitamin D supplementation did not show any effects on the expression of pyruvate dehydrogenase kinase 1 (PDK1) (P=0.37), IRS2 (P=0.90) and lipoprotein (a) [Lp(a)] (P=0.05). CONCLUSION: Our findings confirmed that diabetic HD subjects who received the vitamin D supplementation (for 12 weeks), showed a significant overexpression in the PPAR-γ, AKT, PI3K, IRS1 and GLUT4 genes, and also showed a significant downregulation in the PKC and LDLR genes. Moreover, no effects on PDK1, IRS2 and Lp(a) expression were observed.


Subject(s)
Diabetic Nephropathies/therapy , Gene Expression Regulation/drug effects , Insulin/metabolism , Lipid Metabolism/drug effects , Renal Dialysis , Vitamin D/therapeutic use , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Diabetic Nephropathies/genetics , Dietary Supplements , Double-Blind Method , Enzyme Induction/drug effects , Female , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/genetics , Male , Middle Aged , PPAR gamma/biosynthesis , PPAR gamma/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/biosynthesis , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Signal Transduction , Vitamin D/administration & dosage , Vitamin D/pharmacology , Young Adult
13.
J Med Food ; 22(7): 696-702, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30985232

ABSTRACT

Evidence indicates that indirect inhibitory regulation of glutamatergic transmission, via reducing glutamate release, may induce neuroprotection. The present work was designed to examine whether allicin, a major component of garlic with neuroprotective effects, affected the release of glutamate evoked by 4-aminopyridine in rat cerebrocortical nerve terminals (synaptosomes). Allicin caused a potent inhibition on the release of glutamate evoked by 4-aminopyridine, and this inhibitory effect was abolished in the presence of Ca2+-free medium and vesicular transporter inhibitor. Allicin decreased the 4-aminopyridine-evoked elevation of intrasynaptosomal Ca2+ levels, but had no effect on the synaptosomal plasma membrane potential. The allicin-mediated inhibition of glutamate release was prevented by the N- and P/Q-type channel blocker and the protein kinase C (PKC) inhibitor, but was not affected by the intracellular Ca2+-release inhibitors, mitogen-activated protein kinase inhibitor, and protein kinase A inhibitor. Western blotting data also showed that allicin significantly reduced the phosphorylation of PKC. Together, these data indicate that in rat cerebrocortical nerve terminals, allicin depresses glutamate release and appears to decrease N- and P/Q-type Ca2+ channel and PKC activity.


Subject(s)
Calcium/metabolism , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Nerve Endings/metabolism , Protein Kinase C/metabolism , Sulfinic Acids/pharmacology , Animals , Cerebral Cortex/drug effects , Disulfides , Male , Nerve Endings/drug effects , Neuroprotective Agents/pharmacology , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism
14.
Int J Mol Sci ; 20(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30823645

ABSTRACT

Maternal diet modifies epigenetic programming in offspring, a potentially critical factor in the immune dysregulation of modern societies. We previously found that prenatal fish oil supplementation affects neonatal T-cell histone acetylation of genes implicated in adaptive immunity including PRKCZ, IL13, and TBX21. In this study, we measured H3 and H4 histone acetylation levels by chromatin immunoprecipitation in 173 term placentas collected in the prospective birth cohort, ALADDIN, in which information on lifestyle and diet is thoroughly recorded. In anthroposophic families, regular olive oil usage during pregnancy was associated with increased H3 acetylation at FOXP3 (p = 0.004), IL10RA (p = 0.008), and IL7R (p = 0.007) promoters, which remained significant after adjustment by offspring gender. Furthermore, maternal fish consumption was associated with increased H4 acetylation at the CD14 gene in placentas of female offspring (p = 0.009). In conclusion, prenatal olive oil intake can affect placental histone acetylation in immune regulatory genes, confirming previously observed pro-acetylation effects of olive oil polyphenols. The association with fish consumption may implicate ω-3 polyunsaturated fatty acids present in fish oil. Altered histone acetylation in placentas from mothers who regularly include fish or olive oil in their diets could influence immune priming in the newborn.


Subject(s)
Fish Oils/pharmacology , Histones/metabolism , Maternal Nutritional Physiological Phenomena , Olive Oil/pharmacology , Placenta/metabolism , Protein Processing, Post-Translational , Acetylation , Female , Fish Oils/administration & dosage , Fish Oils/metabolism , Fish Products , Humans , Immunity, Innate/genetics , Interleukin-13/genetics , Interleukin-13/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Olive Oil/administration & dosage , Placenta/drug effects , Pregnancy , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
15.
Fish Shellfish Immunol ; 87: 590-599, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30738864

ABSTRACT

Complementary (c)DNA encoding novel protein kinase C (PKC) messenger (m)RNA of the white shrimp Litopenaeus vannamei, consisted of 2454-bp cDNA containing an open reading frame (ORF) of 2232 bp, belonging to the novel (n)PKC family of proteins characterized by their containing two phorbol ester/diacylglycerol-binding domains (C1 domain), a C2 domain, and a catalytic domain of the serine/threonine kinase, designated LvnPKC. A comparison of amino acid sequences showed that LvnPKC was closely related to arthropod nPKC. LvnPKC cDNA was detected in all tested tissues with a real-time PCR including the hepatopancreas, gills, muscles, subcuticular epithelium, abdominal nerve, thoracic nerve, brain, the stomach, heart, and especially in hemocytes and the intestines. Moreover, significantly upregulated LvnPKC expression was only observed in the eyestalk, brain, and hepatopancreas of shrimp transferred from 28 °C to 18 °C for 30 min. Induction of LvnPKC expression in hemocytes of L. vannamei injected with Vibrio alginolyticus at 105 cfu shrimp-1 was detected earlier than in those injected with 103 cfu shrimp-1. Shrimp received LvnPKC-dsRNA for 1 days specifically depleted the expression of LvnPKC mRNA in hemocytes compared those of diethylpyrocarbonate water treatment. After that, significantly decreased expressions of lipopolysaccharide - and ß-1,3-glucan-binding protein, prophenoloxidase-activating enzyme, peroxinectin, prophenoloxidase I, and prophenoloxidase II in the prophenoloxidase-activating system; lysozyme and cytosolic manganese superoxide dismutase and mitochondrial manganese superoxide dismutase in the antioxidant system were observed. We therefore concluded that LvnPKC is involved in immune defense of L. vannamei exposed to hypothermal stress or infected with V. alginolyticus.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Penaeidae/genetics , Penaeidae/immunology , Protein Kinase C/genetics , Protein Kinase C/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Base Sequence , Gene Expression Profiling , Immunocompetence , Phylogeny , Protein Kinase C/chemistry , Vibrio alginolyticus/physiology
16.
EMBO J ; 37(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30389661

ABSTRACT

Nutrient overload in combination with decreased energy dissipation promotes obesity and diabetes. Obesity results in a hormonal imbalance, which among others activates G protein-coupled receptors utilizing diacylglycerol (DAG) as secondary messenger. Protein kinase D1 (PKD1) is a DAG effector, which integrates multiple nutritional and hormonal inputs, but its physiological role in adipocytes is unknown. Here, we show that PKD1 promotes lipogenesis and suppresses mitochondrial fragmentation, biogenesis, respiration, and energy dissipation in an AMP-activated protein kinase (AMPK)-dependent manner. Moreover, mice lacking PKD1 in adipocytes are resistant to diet-induced obesity due to elevated energy expenditure. Beiging of adipocytes promotes energy expenditure and counteracts obesity. Consistently, deletion of PKD1 promotes expression of the ß3-adrenergic receptor (ADRB3) in a CCAAT/enhancer binding protein (C/EBP)-α- and δ-dependent manner, which leads to the elevated expression of beige markers in adipocytes and subcutaneous adipose tissue. Finally, deletion of PKD1 in adipocytes improves insulin sensitivity and ameliorates liver steatosis. Thus, depletion of PKD1 in adipocytes increases energy dissipation by several complementary mechanisms and might represent an attractive strategy to treat obesity and its related complications.


Subject(s)
Adipocytes/metabolism , Adiposity , Energy Metabolism , Fatty Liver/metabolism , Obesity/metabolism , Protein Kinase C/metabolism , Subcutaneous Fat/metabolism , 3T3-L1 Cells , Adipocytes/pathology , Animals , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Female , Humans , Male , Mice , Mice, Mutant Strains , Obesity/genetics , Obesity/pathology , Protein Kinase C/genetics , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Second Messenger Systems/genetics , Subcutaneous Fat/physiology
17.
J Neurosci ; 38(49): 10489-10504, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30355630

ABSTRACT

Mechanical allodynia, a widespread pain symptom that still lacks effective therapy, is associated with the activation of a dorsally directed polysynaptic circuit within the spinal dorsal horn (SDH) or medullary dorsal horn (MDH), whereby tactile inputs into deep SDH/MDH can gain access to superficial SDH/MDH, eliciting pain. Inner lamina II (IIi) interneurons expressing the γ isoform of protein kinase C (PKCγ+) are key elements for allodynia circuits, but how they operate is still unclear. Combining behavioral, ex vivo electrophysiological, and morphological approaches in an adult rat model of facial inflammatory pain (complete Freund's adjuvant, CFA), we show that the mechanical allodynia observed 1 h after CFA injection is associated with the following (1) sensitization (using ERK1/2 phosphorylation as a marker) and (2) reduced dendritic arborizations and enhanced spine density in exclusively PKCγ+ interneurons, but (3) depolarized resting membrane potential (RMP) in all lamina IIi PKCγ+/PKCγ- interneurons. Blocking MDH 5HT2A receptors (5-HT2AR) prevents facial mechanical allodynia and associated changes in the morphology of PKCγ+ interneurons, but not depolarized RMP in lamina IIi interneurons. Finally, activation of MDH 5-HT2AR in naive animals is enough to reproduce the behavioral allodynia and morphological changes in PKCγ+ interneurons, but not the electrophysiological changes in lamina IIi interneurons, induced by facial inflammation. This suggests that inflammation-induced mechanical allodynia involves strong morphological reorganization of PKCγ+ interneurons via 5-HT2AR activation that contributes to open the gate for transmission of innocuous mechanical inputs to superficial SDH/MDH pain circuitry. Preventing 5-HT2AR-induced structural plasticity in PKCγ+ interneurons might represent new avenues for the specific treatment of inflammation-induced mechanical hypersensitivity.SIGNIFICANCE STATEMENT Inflammatory or neuropathic pain syndromes are characterized by pain hypersensitivity such as mechanical allodynia (pain induced by innocuous mechanical stimuli). It is generally assumed that mechanisms underlying mechanical allodynia, because they are rapid, must operate at only the level of functional reorganization of spinal or medullary dorsal horn (MDH) circuits. We discovered that facial inflammation-induced mechanical allodynia is associated with rapid and strong structural remodeling of specifically interneurons expressing the γ isoform of protein kinase C (PKCγ) within MDH inner lamina II. Moreover, we elucidated a 5-HT2A receptor to PKCγ/ERK1/2 pathway leading to the behavioral allodynia and correlated morphological changes in PKCγ interneurons. Therefore, descending 5-HT sensitize PKCγ interneurons, a putative "gate" in allodynia circuits, via 5-HT2A receptor-induced structural reorganization.


Subject(s)
Gene Expression Regulation, Enzymologic , Hyperalgesia/metabolism , Interneurons/metabolism , Protein Kinase C/biosynthesis , Receptor, Serotonin, 5-HT2A/metabolism , Touch/physiology , Animals , Facial Pain/metabolism , Facial Pain/pathology , Hyperalgesia/genetics , Hyperalgesia/pathology , Inflammation/metabolism , Inflammation/pathology , Interneurons/pathology , Male , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley
18.
Molecules ; 23(7)2018 06 23.
Article in English | MEDLINE | ID: mdl-29937497

ABSTRACT

Background: A variety of causative factors are involved in the initiation of diabetic retinopathy (DR). Current antidiabetic therapies are expensive and not easily accessible by the public. Furthermore, the use of multiple synthetic drugs leads to severe side effects, which worsen the diabetic patient's condition. Medicinal plants and their derived phytochemicals are considered safe and effective treatment and their consumption can reduce the DR risk. In this article, we discuss a variety of medicinal plants, and their noteworthy bio-active constituents, that will be utilized as target based therapeutic strategies for DR. Methods: A broad-spectrum study was conducted using published English works in various electronic databases including Science Direct, PubMed, Scopus, and Google Scholar. Results: Targeting the multiple pathological factors including ROS, AGEs formation, hexosamine flux, PARP, PKC, and MAPK activation through variety of bioactive constituents in medicinal plants, diabetes progression can be delayed with improved loss of vision. Conclusions: Data reveals that traditional herbs and their prominent bioactive components control and normalize pathological cellular factors involved in DR progression. Therefore, studies should be carried out to explore the protective retinopathy effects of medicinal plants using experimental animal and humans models.


Subject(s)
Diabetic Retinopathy/drug therapy , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Phytochemicals/pharmacology , Phytotherapy/methods , Plants, Medicinal/chemistry , Animals , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/metabolism , Hexosamines/antagonists & inhibitors , Hexosamines/metabolism , Humans , Hypoglycemic Agents/isolation & purification , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Phytochemicals/isolation & purification , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology
19.
Nutrients ; 10(4)2018 Apr 06.
Article in English | MEDLINE | ID: mdl-29642372

ABSTRACT

Glioblastoma multiforme (GBM) is a highly infiltrative brain cancer with a dismal prognosis. High levels of brain fatty acid binding protein (B-FABP) are associated with increased migration/infiltration in GBM cells, with a high ratio of arachidonic acid (AA) to docosahexaenoic acid (DHA) driving B-FABP-mediated migration. Since several protein kinase Cs (PKCs) are overexpressed in GBM and linked to migration, we explored a possible relationship between B-FABP and levels/activity of different PKCs, as a function of AA and DHA supplementation. We report that ectopic expression of B-FABP in U87 cells alters the levels of several PKCs, particularly PKCζ. Upon analysis of PKCζ RNA levels in a panel of GBM cell lines and patient-derived GBM neurospheres, we observed a trend towards moderate positive correlation (r = 0.624, p = 0.054) between B-FABP and PKCζ RNA levels. Analysis of PKC activity in U87 GBM cells revealed decreased typical PKC activity (23.4%) in B-FABP-expressing cells compared with nonexpressing cells, with no difference in novel and atypical PKC activities. AA and DHA modulated both conventional and atypical PKC activities in a B-FABP-dependent manner, but had no effect on novel PKC activity. These results suggest that conventional and atypical PKCs are potential downstream effectors of B-FABP/fatty acid-mediated alterations in GBM growth properties.


Subject(s)
Arachidonic Acid/pharmacology , Brain Neoplasms/drug therapy , Docosahexaenoic Acids/pharmacology , Fatty Acid-Binding Protein 7/metabolism , Glioblastoma/drug therapy , Protein Kinase C/metabolism , Tumor Suppressor Proteins/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Protein Kinase C/genetics , Signal Transduction/drug effects
20.
J Med Food ; 21(5): 462-468, 2018 May.
Article in English | MEDLINE | ID: mdl-29341802

ABSTRACT

This study investigated the effects of Portulaca oleracea L. extract on glucose uptake in 3T3-L1 adipocytes. P. oleracea extract (POE) markedly enhanced glucose uptake, which was caused by increased GLUT4 expression at the plasma membrane (PM) in 3T3-L1 adipocytes. This increase in PM-GLUT4 expression was associated with insulin receptor substrate-1 (IRS-1) phosphorylation, phosphatidylinositol 3-kinase (PI3K) activation, and Akt phosphorylation, and finally, enhanced intracellular glucose uptake. POE was not associated with protein kinase C (PKC)λ/ζ phosphorylation in the insulin signaling pathway, but did promote 5'-AMP-activated kinase (AMPK) phosphorylation. Increased glucose uptake through POE was inhibited through treating with the PI3K inhibitor or AMPK inhibitor in 3T3-L1 adipocytes. This result suggested that POE may enhance glucose uptake by stimulating GLUT4 translocation to the PM through activating the PI3K and AMPK pathway in 3T3-L1 adipocytes.


Subject(s)
Carbohydrate Metabolism/drug effects , Cell Membrane/drug effects , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Plant Extracts/pharmacology , Portulaca/chemistry , 3T3-L1 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Androstadienes/pharmacology , Animals , Cell Membrane/metabolism , Glucose Transporter Type 4/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL