Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 516
Filtrar
1.
FASEB J ; 38(8): e23590, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38656553

RESUMO

Studies have suggested that microglial IL-6 modulates inflammatory pain; however, the exact mechanism of action remains unclear. We therefore hypothesized that PKCε and MEG2 competitively bind to STAT3 and contribute to IL-6-mediated microglial hyperalgesia during inflammatory pain. Freund's complete adjuvant (FCA) and lipopolysaccharide (LPS) were used to induce hyperalgesia model mice and microglial inflammation. Mechanical allodynia was evaluated using von Frey tests in vivo. The interaction among PKCε, MEG2, and STAT3 was determined using ELISA and immunoprecipitation assay in vitro. The PKCε, MEG2, t-STAT3, pSTAT3Tyr705, pSTAT3Ser727, IL-6, GLUT3, and TREM2 were assessed by Western blot. IL-6 promoter activity and IL-6 concentration were examined using dual luciferase assays and ELISA. Overexpression of PKCε and MEG2 promoted and attenuated inflammatory pain, accompanied by an increase and decrease in IL-6 expression, respectively. PKCε displayed a stronger binding ability to STAT3 when competing with MEG2. STAT3Ser727 phosphorylation increased STAT3 interaction with both PKCε and MEG2. Moreover, LPS increased PKCε, MEG2, pSTAT3Tyr705, pSTAT3Ser727, IL-6, and GLUT3 levels and decreased TREM2 during microglia inflammation. IL-6 promoter activity was enhanced or inhibited by PKCε or MEG2 in the presence of STAT3 and LPS stimulation, respectively. In microglia, overexpression of PKCε and/or MEG2 resulted in the elevation of tSTAT3, pSTAT3Tyr705, pSTAT3Ser727, IL-6, and TREM2, and the reduction of GLUT3. PKCε is more potent than MEG2 when competitively binding to STAT3, displaying dual modulatory effects of IL-6 production, thus regulating the GLUT3 and TREM2 in microglia during inflammatory pain sensation.


Assuntos
Hiperalgesia , Inflamação , Interleucina-6 , Microglia , Proteína Quinase C-épsilon , Fator de Transcrição STAT3 , Animais , Masculino , Camundongos , Adjuvante de Freund , Hiperalgesia/metabolismo , Inflamação/metabolismo , Interleucina-6/metabolismo , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Dor/metabolismo , Fosforilação , Ligação Proteica , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C-épsilon/genética , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Fator de Transcrição STAT3/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo
2.
Biotech Histochem ; 98(7): 501-507, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37501577

RESUMO

Glioma is the most common type of primary brain tumor; it exhibits great invasive capacity, morbidity and mortality. Protein kinase Cε (PKCε), a serine/threonine kinase, contributes to the development and progression of many cancers. We investigated whether knockdown of PKCε could affect the mitochondrial membrane potential of human glioma cell lines, U251 and U87, and the growth of U251 cell-derived tumors in nude mice. We found that the expression of PKCε was greater in human glioma tissues than in human normal brain tissues. Knockdown of PKCε reduced mitochondrial membrane potential in U251 and U87 cells. Knockdown of PKCε also suppressed the growth of tumors derived from U251 cells and induced apoptosis of U251 cells in vivo. Our findings indicate that PKCε is important for development and progression of glioma and may be a potential therapeutic target for glioma treatment.


Assuntos
Glioma , Proteína Quinase C-épsilon , Animais , Camundongos , Humanos , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C-épsilon/farmacologia , Camundongos Nus , Potencial da Membrana Mitocondrial , Proliferação de Células , Glioma/genética , Glioma/tratamento farmacológico , Glioma/metabolismo , Apoptose , Linhagem Celular Tumoral
3.
Exp Cell Res ; 422(1): 113427, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36400183

RESUMO

Protein kinase C epsilon (PKCε) belongs to a family of serine/threonine kinases that control cell proliferation, differentiation and survival. Aberrant PKCε activation and overexpression is a frequent feature of numerous cancers. However, its role in regulation of lipid metabolism in cancer cells remains elusive. Here we report a novel function of PKCε in regulating of prostate cancer cell proliferation by modulation of PKM2-mediated de novo lipogenesis. We show that PKCε promotes de novo lipogenesis and tumor cell proliferation via upregulation of lipogenic enzymes and lipid contents in prostate cancer cells. Mechanistically, PKCε interacts with NABD (1-388) domain of C-terminal deletion on pyruvate kinase isoform M2 (PKM2) and enhances the Tyr105 phosphorylation of PKM2, leading to its nuclear localization. Moreover, forced expression of mutant Tyr105 (Y105F) or PKM2 inhibition suppressed de novo lipogenesis and cell proliferation induced by overexpression of PKCε in prostate cancer cells. In a murine tumor model, inhibitor of PKM2 antagonizes lipogenic enzymes expression and prostate cancer growth induced by overexpression of PKCε in vivo. These data indicate that PKCε is a critical regulator of de novo lipogenesis, which may represent a potential therapeutic target for the treatment of prostate cancer.


Assuntos
Neoplasias da Próstata , Proteína Quinase C-épsilon , Animais , Humanos , Masculino , Camundongos , Linhagem Celular Tumoral , Lipogênese/genética , Fosforilação/fisiologia , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/metabolismo , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Piruvato Quinase/genética , Piruvato Quinase/metabolismo
4.
Int J Mol Sci ; 23(18)2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36142751

RESUMO

Exercise induces cardioprotection against myocardial infarction, despite obesity, by restoring pro-survival pathways and increasing resistance of mitochondrial permeability transition pore (mPTP) opening at reperfusion. Among the mechanisms involved in the inactivation of these pathways, oxysterols appear interesting. Thus, we investigated the influence of regular exercise on the reperfusion injury salvage kinase (RISK) pathway, oxysterols, and mitochondria, in the absence of ischemia-reperfusion. We also studied 7ß-hydroxycholesterol (7ßOH) concentration (mass spectrometry) in human lean and obese subjects. Wild-type (WT) and obese (ob/ob) mice were assigned to sedentary conditions or regular treadmill exercise. Exercise significantly increased Akt phosphorylation, whereas 7ßOH concentration was reduced. Moreover, exercise induced the translocation of PKCε from the cytosol to mitochondria. However, exercise did not affect the calcium concentration required to open mPTP in the mitochondria, neither in WT nor in ob/ob animals. Finally, human plasma 7ßOH concentration was consistent with observations made in mice. In conclusion, regular exercise enhanced the RISK pathway by increasing kinase phosphorylation and PKCε translocation and decreasing 7ßOH concentration. This activation needs the combination with stress conditions, i.e., ischemia-reperfusion, in order to inhibit mPTP opening at the onset of reperfusion. The human findings suggest 7ßOH as a candidate marker for evaluating cardiovascular risk factors in obesity.


Assuntos
Traumatismo por Reperfusão Miocárdica , Oxisteróis , Animais , Humanos , Camundongos , Cálcio/metabolismo , Camundongos Obesos , Mitocôndrias Cardíacas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Traumatismo por Reperfusão Miocárdica/metabolismo , Obesidade/metabolismo , Oxisteróis/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia
5.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-35805984

RESUMO

Hypobaric hypoxia is a condition that occurs at high altitudes (>2500 m) where the partial pressure of gases, particularly oxygen (PO2), decreases. This condition triggers several physiological and molecular responses. One of the principal responses is pulmonary vascular contraction, which seeks to optimize gas exchange under this condition, known as hypoxic pulmonary vasoconstriction (HPV); however, when this physiological response is exacerbated, it contributes to the development of high-altitude pulmonary hypertension (HAPH). Increased levels of zinc (Zn2+) and oxidative stress (known as the "ROS hypothesis") have been demonstrated in the vasoconstriction process. Therefore, the aim of this review is to determine the relationship between molecular pathways associated with altered Zn2+ levels and oxidative stress in HPV in hypobaric hypoxic conditions. The results indicate an increased level of Zn2+, which is related to increasing mitochondrial ROS (mtROS), alterations in nitric oxide (NO), metallothionein (MT), zinc-regulated, iron-regulated transporter-like protein (ZIP), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-induced protein kinase C epsilon (PKCε) activation in the development of HPV. In conclusion, there is an association between elevated Zn2+ levels and oxidative stress in HPV under different models of hypoxia, which contribute to understanding the molecular mechanism involved in HPV to prevent the development of HAPH.


Assuntos
Infecções por Papillomavirus , Vasoconstrição , Doença da Altitude , Humanos , Hipertensão Pulmonar , Hipóxia/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo , Infecções por Papillomavirus/metabolismo , Proteína Quinase C-épsilon/metabolismo , Artéria Pulmonar/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Vasoconstrição/fisiologia , Zinco/metabolismo
6.
Int Immunopharmacol ; 108: 108905, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35729836

RESUMO

Studies have confirmed that the heart is the main target organ of lipopolysaccharide (LPS) attacks, and 14-3-3γ and protein kinase C epsilon (PKCε) are the endogenous protective proteins. Puerarin (Pue) is the major bioactive ingredient isolated from the root of Pueraria lobata. It possesses many pharmacological properties, which has been widely used in the treatment and adjuvant therapy of cardio- and cerebrovascular diseases and cancer, etc. The study intended to explore the effects and mechanism of Pue pretreatment to protect myocardium against LPS injury. Adult mice and primary cultured neonatal rat cardiomyocytes were pretreated with Pue, and the injury model was made with LPS. Results showed that Pue pretreatment alleviated LPS-induced injury, as demonstrated by increased cell viability, decreased LDH activity and apoptosis, inhibited excess oxidative stress and the inflammatory cytokine release, and maintained mitochondrial function. Furthermore, Pue pretreatment upregulated 14-3-3γ expression, interacted with PKCε, which was phosphorylated and impelled migration to mitochondria, and then activated adaptive autophagy and protected the myocardium. However, pAD/14-3-3γ-shRNA or 3-MA (an autophagy inhibitor) could weaken the above effects of Pue pretreatment. Together, Pue pretreatment could activate adaptive autophagy by the 14-3-3γ/PKCε pathway and protect the myocardium against LPS injury.


Assuntos
Traumatismos Cardíacos , Isoflavonas , Animais , Apoptose , Autofagia , Isoflavonas/farmacologia , Isoflavonas/uso terapêutico , Lipopolissacarídeos/farmacologia , Camundongos , Miócitos Cardíacos/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C-épsilon/farmacologia , Ratos , Ratos Sprague-Dawley
7.
Biomolecules ; 12(3)2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35327585

RESUMO

Most individuals diagnosed with alcohol use disorders smoke cigarettes. Large concentrations of malondialdehyde and acetaldehyde are found in lungs co-exposed to cigarette smoke and alcohol. Aldehydes directly injure lungs and form aldehyde protein adducts, impacting epithelial functions. Recently, 2-(3-Amino-6-chloroquinolin-2-yl)propan-2-ol (ADX-102) was developed as an aldehyde-trapping drug. We hypothesized that aldehyde-trapping compounds are protective against lung injury derived from cigarette smoke and alcohol co-exposure. To test this hypothesis, we pretreated mouse ciliated tracheal epithelial cells with 0-100 µM of ADX-102 followed by co-exposure to 5% cigarette smoke extract and 50 mM of ethanol. Pretreatment with ADX-102 dose-dependently protected against smoke and alcohol induced cilia-slowing, decreases in bronchial epithelial cell wound repair, decreases in epithelial monolayer resistance, and the formation of MAA adducts. ADX-102 concentrations up to 100 µM showed no cellular toxicity. As protein kinase C (PKC) activation is a known mechanism for slowing cilia and wound repair, we examined the effects of ADX-102 on smoke and alcohol induced PKC epsilon activity. ADX-102 prevented early (3 h) activation and late (24 h) autodownregulation of PKC epsilon in response to smoke and alcohol. These data suggest that reactive aldehydes generated from cigarette smoke and alcohol metabolism may be potential targets for therapeutic intervention to reduce lung injury.


Assuntos
Alcoolismo , Fumar Cigarros , Lesão Pulmonar , Alcoolismo/metabolismo , Aldeídos/metabolismo , Aminoquinolinas , Animais , Etanol/toxicidade , Pulmão/metabolismo , Camundongos , Proteína Quinase C-épsilon/metabolismo , Nicotiana
8.
Mediators Inflamm ; 2022: 2782080, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35125963

RESUMO

METHODS: A rat hyperalgesia model was induced using an intraplantar injection of Freund's complete adjuvant (FCA) or an intrathecal injection of IL-6. Mechanical allodynia was evaluated using von Frey filament tests after intrathecal injections of T-5224 (c-Fos/AP-1 inhibitor), minocycline (Mino, a specific microglia inhibitor), L-2-aminoadipic acid (LAA, an astroglial toxin), PKCε inhibitor peptide, APTSTAT3-9R (STAT3 inhibitor), or anti-IL-6 antibody. The c-Fos, GFAP, Iba-1, PKCε, STAT3, pSTAT3Tyr705 and pSTAT3Ser727, and IL-6 expression at the spinal cord level was assessed by Western blot analysis. The interactive effects of PKCε and STAT3 were determined using immunofluorescence staining and immunoprecipitation in vivo and in vitro. Interleukin-6 promoter activity was examined using luciferase assays. RESULTS: T-5224, Mino, and LAA attenuated FCA- or IL-6-mediated inflammatory pain, with a decrease in c-Fos, GFAP, Iba-1, PKCε, and IL-6 expression. PKCε inhibitor peptide and APTSTAT3-9R reversed FCA-induced nociceptive behavior, while decreasing pSTAT3Ser727, IL-6, c-Fos, GFAP, and Iba-1 expression and PKCε and STAT3 coexpression. Interleukin-6 promoter activity increased in the presence of PKCε and STAT3. The interaction with PKCε increased on phosphorylating STAT3 at Ser727 but not at Tyr705. CONCLUSION: STAT3 phosphorylation at Ser 727 and the interaction with PKCε contribute to hyperalgesia via the IL-6-mediated signaling pathway, thus regulating neuron-glia crosstalk during inflammatory pain.


Assuntos
Hiperalgesia , Interleucina-6 , Animais , Hiperalgesia/metabolismo , Interleucina-6/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fosforilação , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C-épsilon/farmacologia , Ratos , Medula Espinal/metabolismo
9.
Hum Mol Genet ; 31(13): 2236-2261, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35137073

RESUMO

Novel protein kinase C (nPKC) family member, protein kinase C epsilon (PKCε) is an AGC kinase superfamily member. It is associated with neurological and metabolic diseases as well as human cancers. No study so far has been conducted to identify genetic variations and their effect on PKCε folding and functioning. The present study aimed to identify mutational hotspots in PKCε and disease-causing non-synonymous variants (nsSNPs) along with the investigation of nsSNP impact on protein dynamics. Twenty-nine in silico tools were applied to determine nsSNP deleteriousness, their impact on protein dynamics and disease association, along with the prediction of PKCε post-translational modification (PTM) sites. The present study's outcomes indicated that most nsSNPs were concentrated in the PKCε hinge region and C-terminal tail. Most pathogenic variants mapped to the kinase domain. Regulatory domain variants influenced PKCε interaction with molecular players whereas kinase domain variants were predicted to impact its phosphorylation pattern and protein-protein interactions. Most PTM sites were mapped to the hinge region. PKCε nsSNPs have an association with oncogenicity and its expression dysregulation is responsible for poor overall survival. Understanding nsSNP structural impact is a primary step necessary for delineating the relationship of genetic level differences with protein phenotype. The obtained knowledge can eventually help in disease diagnosis and therapy design.


Assuntos
Proteína Quinase C-épsilon , Proteínas , Mutação , Fenótipo , Fosforilação , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Proteínas/genética
10.
Neurosci Lett ; 766: 136348, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785308

RESUMO

Pregabalin has been increasingly used in recent years, and becoming a first-line medication for the clinical treatment of neuropathic pain. However, the mechanisms underlying pregabalin-induced neuropathic pain alleviation remain unclear. In this study, we aimed to investigate whether PKC epsilon (PKCε)/ transient receptor potential vanilloid subtype 1(TRPV1) signaling pathway participated in pregabalin-induced analgesia during treatment of neuropathic pain using rat models of spared nerve injury (SNI). The left hind paw withdrawal mechanical thresholds (PWMT) of rats were measured preoperatively one day before and on day 1, 4, 7 and 14 after surgery. On day 7 after SNI surgery, the rats received ligation operation were administrated with pregabalin intraperitoneally and were intrathecally injected with PKC Inhibitor BIM Ⅰ or PKC agonist PMA for seven consecutive days, IL-1ß and IL-6 expression levels in the spinal cord of rats were then assessed. Furthermore, we analyzed the PKCε, TRPV1, pTRPV1 and Glial fibrillary acidic protein (GFAP) protein levels and the expression of reactive astrocytes and the PKCε, TRPV1 and pTRPV1 positive cells on day 14 after SNI. Our findings indicated that pregabalin could relieve neuropathic pain to a certain extent by suppressing the PKCε/TRPV1 signaling pathway and inhibiting inflammatory processes in the spinal cord.


Assuntos
Analgésicos/farmacologia , Neuralgia/metabolismo , Pregabalina/farmacologia , Proteína Quinase C-épsilon/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos
11.
Int J Mol Sci ; 22(20)2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34681669

RESUMO

Aging attenuates cardiac tolerance to ischemia/reperfusion (I/R) associated with defects in protective cell signaling, however, the onset of this phenotype has not been completely investigated. This study aimed to compare changes in response to I/R and the effects of remote ischemic preconditioning (RIPC) in the hearts of younger adult (3 months) and mature adult (6 months) male Wistar rats, with changes in selected proteins of protective signaling. Langendorff-perfused hearts were exposed to 30 min I/120 min R without or with prior three cycles of RIPC (pressure cuff inflation/deflation on the hind limb). Infarct size (IS), incidence of ventricular arrhythmias and recovery of contractile function (LVDP) served as the end points. In both age groups, left ventricular tissue samples were collected prior to ischemia (baseline) and after I/R, in non-RIPC controls and in RIPC groups to detect selected pro-survival proteins (Western blot). Maturation did not affect post-ischemic recovery of heart function (Left Ventricular Developed Pressure, LVDP), however, it increased IS and arrhythmogenesis accompanied by decreased levels and activity of several pro-survival proteins and by higher levels of pro-apoptotic proteins in the hearts of elder animals. RIPC reduced the occurrence of reperfusion-induced ventricular arrhythmias, IS and contractile dysfunction in younger animals, and this was preserved in the mature adults. RIPC did not increase phosphorylated protein kinase B (p-Akt)/total Akt ratio, endothelial nitric oxide synthase (eNOS) and protein kinase Cε (PKCε) prior to ischemia but only after I/R, while phosphorylated glycogen synthase kinase-3ß (GSK3ß) was increased (inactivated) before and after ischemia in both age groups coupled with decreased levels of pro-apoptotic markers. We assume that resistance of rat heart to I/R injury starts to already decline during maturation, and that RIPC may represent a clinically relevant cardioprotective intervention in the elder population.


Assuntos
Precondicionamento Isquêmico Miocárdico , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Envelhecimento , Animais , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Hemodinâmica , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar
12.
Cells ; 10(10)2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34685716

RESUMO

Mitochondria, abundant organelles in high energy demand cells such as cardiomyocytes, can determine cell death or survival by regulating the opening of mitochondrial permeability transition pore, mPTP. We addressed the hypothesis that the growth factor FGF2, known to reside in intracellular locations, can directly influence mitochondrial susceptibility to mPTP opening. Rat cardiac subsarcolemmal (SSM) or interfibrillar (IFM) mitochondrial suspensions exposed directly to rat 18 kDa low molecular weight (Lo-) FGF2 isoform displayed increased resistance to calcium overload-induced mPTP, measured spectrophotometrically as "swelling", or as cytochrome c release from mitochondria. Inhibition of mitochondrial protein kinase C epsilon abrogated direct Lo-FGF2 mito-protection. Exposure to the rat 23 kDa high molecular weight (Hi) FGF2 isoform promoted cytochrome c release from SSM and IFM under nonstressed conditions. The effect of Hi-FGF2 was prevented by mPTP inhibitors, pre-exposure to Lo-FGF2, and okadaic acid, a serine/threonine phosphatase inhibitor. Western blotting and immunoelectron microscopy pointed to the presence of immunoreactive FGFR1 in cardiac mitochondria in situ. The direct mito-protective effect of Lo-FGF2, as well as the deleterious effect of Hi-FGF2, were prevented by FGFR1 inhibitors and FGFR1 neutralizing antibodies. We propose that intracellular FGF2 isoforms can modulate mPTP opening by interacting with mito-FGFR1 and relaying isoform-specific intramitochondrial signal transduction.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Mitocôndrias Cardíacas/metabolismo , Poro de Transição de Permeabilidade Mitocondrial/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Cálcio/metabolismo , Citocromos c/metabolismo , Masculino , Mitocôndrias Cardíacas/ultraestrutura , Isoformas de Proteínas/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores
13.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34433666

RESUMO

Increasing attention has been paid to roles of tripartite motif-containing (TRIM) family proteins in cancer biology, often functioning as E3 ubiquitin ligases. In the present study, we focus on a contribution of TRIM47 to breast cancer biology, particularly to endocrine therapy resistance, which is a major clinical problem in breast cancer treatment. We performed immunohistochemical analysis of TRIM47 protein expression in 116 clinical samples of breast cancer patients with postoperative endocrine therapy using tamoxifen. Our clinicopathological study showed that higher immunoreactivity scores of TRIM47 were significantly associated with higher relapse rate of breast cancer patients (P = 0.012). As functional analyses, we manipulated TRIM47 expression in estrogen receptor-positive breast cancer cells MCF-7 and its 4-hydroxytamoxifen (OHT)-resistant derivative OHTR, which was established in a long-term culture with OHT. TRIM47 promoted both MCF-7 and OHTR cell proliferation. MCF-7 cells acquired tamoxifen resistance by overexpressing exogenous TRIM47. We found that TRIM47 enhances nuclear factor kappa-B (NF-κB) signaling, which further up-regulates TRIM47. We showed that protein kinase C epsilon (PKC-ε) and protein kinase D3 (PKD3), known as NF-κB-activating protein kinases, are directly associated with TRIM47 and stabilized in the presence of TRIM47. As an underlying mechanism, we showed TRIM47-dependent lysine 27-linked polyubiquitination of PKC-ε. These results indicate that TRIM47 facilitates breast cancer proliferation and endocrine therapy resistance by forming a ternary complex with PKC-ε and PKD3. TRIM47 and its associated kinases can be a potential diagnostic and therapeutic target for breast cancer refractory to endocrine therapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Resistencia a Medicamentos Antineoplásicos , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Tamoxifeno/uso terapêutico , Proteínas de Transporte/genética , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Células MCF-7 , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon/metabolismo , Estabilidade Proteica , Ubiquitinação
14.
Iran Biomed J ; 25(5): 343-8, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34425652

RESUMO

Background: Alzheimer's disease is one of the neurodegenerative disorders typified by the aggregate of amyloid-ß (Aß) and phosphorylated tau protein. Oxidative stress and neuroinflammation, because of Aß peptides, are strongly involved in the pathophysiology of Alzheimer's disease (AD). Linagliptin shows neuroprotective properties against AD pathological processes through alleviation of neural inflammation and AMPK activation. Methods: We assessed the benefits of linagliptin pretreatment (at 10, 20, and 50 nM concentrations), against Aß1-42 toxicity (20 µM) in SH-SY5Y cells. The concentrations of secreted cytokines, such as TNF-α, IL-6, and IL-1ß, and signaling proteins, including pCREB, Wnt1, and PKCε, were quantified by ELISA. Results: We observed that Aß led to cellular inflammation, which was assessed by measuring inflammatory cytokines (TNF-α, IL-1ß, and IL-6). Moreover, Aß1-42 treatment impaired pCREB, PKCε, and Wnt1 signaling in human SH-SY5Y neuroblastoma cells. Addition of Linagliptin significantly reduced IL-6 levels in the lysates of cells, treated with Aß1-42. Furthermore, linagliptin prevented the downregulation of Wnt1 in Aß1-42-treated cells exposed. Conclusion: The current findings reveal that linagliptin alleviates Aß1-42-induced inflammation in SH-SY5Y cells, probably through the suppression of IL-6 release, and some of its benefits are mediated through the activation of the Wnt1 signaling pathway.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Interleucina-6/metabolismo , Linagliptina/farmacologia , Neuroblastoma/patologia , Fármacos Neuroprotetores/farmacologia , Proteína Wnt1/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Neuroblastoma/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo
15.
Mol Cells ; 44(8): 569-579, 2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34294609

RESUMO

Cyclase-associated protein 2 (CAP2) has been addressed as a candidate biomarker in various cancer types. Previously, we have shown that CAP2 is expressed during multi-step hepatocarcinogenesis; however, its underlying mechanisms in liver cancer cells are not fully elucidated yet. Here, we demonstrated that endoplasmic reticulum (ER) stress induced CAP2 expression, and which promoted migration and invasion of liver cancer cells. We also found that the ER stress-induced CAP2 expression is mediated through activation of protein kinase C epsilon (PKCε) and the promotor binding of activating transcription factor 2 (ATF2). In addition, we further demonstrated that CAP2 expression promoted epithelial-mesenchymal transition (EMT) through activation of Rac1 and ERK. In conclusion, we suggest that ER stress induces CAP2 expression promoting EMT in liver cancers cells. Our results shed light on the novel functions of CAP2 in the metastatic process of liver cancer cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Estresse do Retículo Endoplasmático , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Fator 2 Ativador da Transcrição/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteína Quinase C-épsilon/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
J Neuroinflammation ; 18(1): 106, 2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-33952299

RESUMO

BACKGROUND: Nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2)-induced oxidative stress, including the production of reactive oxygen species (ROS) and hydrogen peroxide, plays a pivotal role in neuropathic pain. Although the activation and plasma membrane translocation of protein kinase C (PKC) isoforms in dorsal root ganglion (DRG) neurons have been implicated in multiple pain models, the interactions between NOX2-induced oxidative stress and PKC remain unknown. METHODS: A spared nerve injury (SNI) model was established in adult male rats. Pharmacologic intervention and AAV-shRNA were applied locally to DRGs. Pain behavior was evaluated by Von Frey tests. Western blotting and immunohistochemistry were performed to examine the underlying mechanisms. The excitability of DRG neurons was recorded by whole-cell patch clamping. RESULTS: SNI induced persistent NOX2 upregulation in DRGs for up to 2 weeks and increased the excitability of DRG neurons, and these effects were suppressed by local application of gp91-tat (a NOX2-blocking peptide) or NOX2-shRNA to DRGs. Of note, the SNI-induced upregulated expression of PKCε but not PKC was decreased by gp91-tat in DRGs. Mechanical allodynia and DRG excitability were increased by ψεRACK (a PKCε activator) and reduced by εV1-2 (a PKCε-specific inhibitor). Importantly, εV1-2 failed to inhibit SNI-induced NOX2 upregulation. Moreover, the SNI-induced increase in PKCε protein expression in both the plasma membrane and cytosol in DRGs was attenuated by gp91-tat pretreatment, and the enhanced translocation of PKCε was recapitulated by H2O2 administration. SNI-induced upregulation of PKCε was blunted by phenyl-N-tert-butylnitrone (PBN, an ROS scavenger) and the hydrogen peroxide catalyst catalase. Furthermore, εV1-2 attenuated the mechanical allodynia induced by H2O2 CONCLUSIONS: NOX2-induced oxidative stress promotes the sensitization of DRGs and persistent pain by increasing the plasma membrane translocation of PKCε.


Assuntos
NADPH Oxidase 2/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Proteína Quinase C-épsilon/metabolismo , Animais , Membrana Celular/metabolismo , Gânglios Espinais/metabolismo , Masculino , Traumatismos dos Nervos Periféricos/metabolismo , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
17.
Mol Biol Cell ; 32(15): 1347-1358, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34010016

RESUMO

The presence of tight junction protein zonula occludens 2 (ZO-2) at the nucleus inhibits the transcription of genes regulated by TEAD transcription factor. Here, we analyzed whether the movement of ZO-2 into the nucleus modulates the nuclear concentration of TEAD. In sparse cultures of ZO-2 knockdown Madin-Darby canine kidney cells, nuclear TEAD was diminished, as in parental cells transfected with a ZO-2 construct without nuclear localization signals, indicating that ZO-2 facilitates the entry of TEAD into the nucleus. Inhibition of nPKCδ in parental cells triggers the interaction between ZO-2 and TEAD at the cytoplasm and facilitates TEAD/ZO-2 complex nuclear importation. Using proximity ligation, immunoprecipitation, and pull-down assays, TEAD/ZO-2 interaction was confirmed. Nuclear TEAD is phosphorylated, and its exit in parental cells is enhanced by activation of a ZO-2 nuclear exportation signal by nPKCε, while the nuclear accumulation of ZO-2 triggered by the mutation of ZO-2 nuclear export signals induces no change in TEAD nuclear concentration. In summary, our results indicate that the movements of ZO-2 in and out of the nucleus modulate the intracellular traffic of TEAD through a process regulated by nPKCδ and ε and provide a novel role of ZO-2 as a nuclear translocator of TEAD.


Assuntos
Núcleo Celular/metabolismo , Células Epiteliais/metabolismo , Fatores de Transcrição de Domínio TEA/metabolismo , Proteína da Zônula de Oclusão-2/metabolismo , Animais , Linhagem Celular , Cães , Células HEK293 , Humanos , Sinais de Localização Nuclear , Fosforilação , Ligação Proteica , Proteína Quinase C-épsilon/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Ratos , Transdução de Sinais
18.
Cytokine ; 142: 155498, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33773907

RESUMO

Activation of CXCR2 by chemokines such as CXCL1 and CXCL2 increases aggressiveness of breast cancer, inducing chemoresistance, hence CXCR2 antagonists are in clinical trials. We previously reported that inhibition of CXCR2 increases MIP-2 (CXCL2), which may inhibit anti-tumoral effects of CXCR2 antagonists. This seems to be due to inhibition of protein kinase C (PKC) by CXCR2 antagonist since specific inhibitor of PKC also enhances MIP-2 secretion. We here examined whether CXCR2 inhibitor also increases KC (CXCL1) secretion, ligand for CXCR2 involved in metastasis and PKC activators can prevent increases in chemokine secretion. We used SB 225002, which is a specific CXCR2 antagonist. The effects of PKC activators that have documented anti-tumoral effects and activates multiple isozymes of PKC such as Ingenol-3-angelate (I3A) and bryostatin-1 were examined here. In addition, FR236924, PKCε selective and 7α-acetoxy-6ß-benzoyloxy-12-O-benzoylroyleanone (Roy-Bz), PKCδ selective activators were also tested. The effects of activators were determined using brain metastatic (4TBM) and heart metastatic (4THM) subset of 4T1 breast carcinoma cells because these aggressive carcinoma cells with cancer stem cell features secrete high levels of KC and MIP-2. Inhibition of CXCR-2 activity increased KC (CXCL1) secretion. PKC activators prevented SB225002-induced increases in KC and MIP-2 secretion. Different activators/modulators induce differential changes in basal and SB225002-induced chemokine secretion as well as cell proliferation and the activators that act on PKCδ and/or PKCε such as bryostatin 1, FR236924 and Roy-Bz are the most effective. These activators alone also decrease cell proliferation or chemokine secretion or both. Given the role of KC and MIP-2 in drug resistance including chemotherapeutics, activators of PKCε and PKCδ may prevent emerging of resistance to CXCR2 inhibitors as well as other chemotherapeutics.


Assuntos
Quimiocinas/metabolismo , Ativadores de Enzimas/farmacologia , Neoplasias Mamárias Animais/enzimologia , Neoplasias Mamárias Animais/patologia , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Receptores de Interleucina-8B/antagonistas & inibidores , Alcanos/farmacologia , Animais , Briostatinas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL2/metabolismo , Ciclopropanos/farmacologia , Diterpenos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Indóis/farmacologia , Camundongos Endogâmicos BALB C , Compostos de Fenilureia/farmacologia , Receptores de Interleucina-8B/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Biofactors ; 47(4): 587-599, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33740285

RESUMO

IR (insulin resistance) in diabetic brain gave rise to the generation of toxic factor Aß42 and axon collapse which were the marker of AD (Alzheimer's disease)-like lesions in the circumstance of diabetes mellitus. But the underling molecular mechanism was not clear. Chronic HGHI (high glucose and high insulin) exposure accelerates IR has been reported in type II diabetes models. Berberine has been shown to promising effect for IR in vitro and in vivo. This study demonstrates the protective effect and the underlying mechanism of berberine on HGHI-induced IR. HGHI-induced cells were used to mimic the hyperinsulinemia resulting in IR. Berberine was used to uncover the mechanisms for the treatment of hyperinsulinemia in IR model. Morris water maze (MWM), PET imaging, CCK8 assay, ELISA assay, glucose kits, microscopy, and western blot analysis were performed to evaluate the protective effects of berberine. Berberine-improved HGHI-induced IR was correlated with the increase of glucose application in neurons. Meanwhile, the expressions of Pi3K, as well as GLUT3, PKCε, and APP were downregulated in the model, while p-IRS Ser307 was upregulated compared with Normal group. Fortunately, these scenes were reversed by berberine administration. Furthermore, berberine decreased GSK3ß Y216 expressions, inhibited the production of oligomer Aß42 and extended neuronal axon. The monomeric berberine treatment improves IR that may be involved in glucose effective application, rectifying the related proteins of the aberrant insulin pathway. Additionally, it suppressed the generation of Aß42 and ameliorated neuron axon damage. Finally, berberine improves DM (diabetes mellitus)-induced cognitive impairment.


Assuntos
Berberina/farmacologia , Disfunção Cognitiva/prevenção & controle , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/farmacologia , Fosfatidilinositol 3-Quinases/genética , Proteína Quinase C-épsilon/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Linhagem Celular Tumoral , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Regulação da Expressão Gênica , Glucose/antagonistas & inibidores , Glucose/farmacologia , Transportador de Glucose Tipo 3/genética , Transportador de Glucose Tipo 3/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Insulina/farmacologia , Resistência à Insulina , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Metformina/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Estreptozocina/administração & dosagem
20.
Small GTPases ; 12(3): 202-208, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-31648598

RESUMO

Oncogenic protein kinase C epsilon (PKCε) promotes the formation of membrane ruffles and motility in non-small cell lung cancer (NSCLC) cells. We found that PKCε is down-regulated when NSCLC cells undergo epithelial-to-mesenchymal transition (EMT) in response to TGF-ß, thus becoming dispensable for migration and invasion in the mesenchymal state. PKCε silencing or inhibition leads to stress fibre formation, suggesting that this kinase negatively regulates RhoA activity. Ruffle formation induced by PKCε activation in the epithelial state is dependent on PI3K, but does not involve the PI3K-dependent Rac-GEFs Ect2, Trio, Vav2 or Tiam1, suggesting alternative Rac-GEFs as mediators of this response. In the proposed model, PKCε acts as a rheostat for Rho GTPases that differs in the epithelial and mesenchymal states.


Assuntos
Citoesqueleto de Actina/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Transição Epitelial-Mesenquimal , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína Quinase C-épsilon/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Apoptose , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteína Quinase C-épsilon/genética , Transdução de Sinais , Células Tumorais Cultivadas , Proteínas rho de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA