Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.462
Filtrar
1.
Int J Mol Sci ; 25(16)2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39201609

RESUMEN

Ocrelizumab (OCR) is a humanized anti-CD20 monoclonal antibody approved for both Relapsing and Primary Progressive forms of Multiple Sclerosis (MS) treatment. OCR is postulated to act via rapid B cell depletion; however, by analogy with other anti-CD20 agents, additional effects can be envisaged, such as on Protein Kinase C (PKC). Hence, this work aims to explore novel potential mechanisms of action of OCR in peripheral blood mononuclear cells from MS patients before and after 12 months of OCR treatment. We first assessed, up-stream, PKCßII and subsequently explored two down-stream pathways: hypoxia-inducible factor 1 alpha (HIF-1α)/vascular endothelial growth factor (VEGF), and human antigen R (HuR)/manganese-dependent superoxide dismutase (MnSOD) and heat shock proteins 70 (HSP70). At baseline, higher levels of PKCßII, HIF-1α, and VEGF were found in MS patients compared to healthy controls (HC); interestingly, the overexpression of this inflammatory cascade was counteracted by OCR treatment. Conversely, at baseline, the content of HuR, MnSOD, and HSP70 was significantly lower in MS patients compared to HC, while OCR administration induced the up-regulation of these neuroprotective pathways. These results enable us to disclose the dual positive action of OCR: anti-inflammatory and neuroprotective. Therefore, in addition to B cell depletion, the effect of OCR on these molecular cascades can contribute to counteracting disease progression.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Esclerosis Múltiple , Proteína Quinasa C beta , Humanos , Femenino , Proteína Quinasa C beta/metabolismo , Masculino , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Persona de Mediana Edad , Factor A de Crecimiento Endotelial Vascular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Superóxido Dismutasa/metabolismo
2.
J Med Chem ; 67(17): 14885-14911, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39151060

RESUMEN

As an obstinate cancer pancreatic cancer (PC) poses a major challenge due to limited treatment options which include resection surgery, radiation therapy, and gemcitabine-based chemotherapy. In cancer cells, protein kinase C ßI (PKCßI) participates in diverse cellular processes, including cell proliferation, invasion, and apoptotic pathways. In the present study, we created a scaffold to develop PKCßI inhibitors using evodiamine-based synthetic molecules. Among the candidate inhibitors, Evo312 exhibited the highest antiproliferative efficacy against PC cells, PANC-1, and acquired gemcitabine-resistant PC cells, PANC-GR. Additionally, Evo312 robustly inhibited PKCßI activity. Mechanistically, Evo312 effectively suppressed the upregulation of PKCßI protein expression, leading to the induction of cell cycle arrest and apoptosis in PANC-GR cells. Furthermore, Evo312 exerted an antitumor activity in a PANC-GR cell-implanted xenograft mouse model. These findings position Evo312 as a promising lead compound for overcoming gemcitabine resistance in PC through novel mechanisms.


Asunto(s)
Antineoplásicos , Apoptosis , Proliferación Celular , Desoxicitidina , Resistencia a Antineoplásicos , Gemcitabina , Neoplasias Pancreáticas , Quinazolinas , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Animales , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/química , Resistencia a Antineoplásicos/efectos de los fármacos , Quinazolinas/farmacología , Quinazolinas/química , Quinazolinas/síntesis química , Ratones , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Ratones Desnudos , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Relación Estructura-Actividad , Ratones Endogámicos BALB C
3.
Life Sci ; 355: 122994, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39163903

RESUMEN

AIMS: In contrast to G protein-coupled receptors or receptor tyrosine kinases, the mechanism underlying ERK activation through nicotine acetylcholine receptors (nAChRs), members of the ligand-gated ion channel family, remains poorly elucidated. This study aimed to delineate the signaling pathway responsible for ERK activation by the α4ß2 nAChR subtype, which is implicated in nicotine addiction and various mental disorders. MATERIALS AND METHODS: Loss-of-function strategies and mutants of arrestin2/PKCßII with distinct functional characteristics were employed to identify the cellular components and processes involved in ERK activation. KEY FINDINGS: ERK activation via α4ß2 nAChR was observed within the nucleus and necessitated the nuclear translocation of arrestin2 and PKCßII, which exhibited mutual augmentation. Activation of PKCßII by α4ß2 nAChR stimulation facilitated the nuclear translocation of arrestin2 by enhancing its interaction with importin ß1. Apart from scaffolding ERK activation in the nucleus, arrestin2, in cooperation with GRK2, facilitated the activation of the Src/Syk/PKCßII signaling cascade, leading to the nuclear entry of PKCßII in a Gßγ-dependent manner. Upon nuclear localization, PKCßII underwent ubiquitination by Mdm2 and interacted with MEK1, resulting in ERK activation. In summary, α4ß2 nAChR-mediated ERK activation in the nucleus involves the nuclear translocation of arrestin2 and PKCßII, which is reciprocally facilitated via positive feedback augmentation. SIGNIFICANCE: As α4ß2 nAChRs play a pivotal role in various cellular processes including drug addiction and mental disorders, our findings will offer insights into understanding the pathogenesis of α4ß2 nAChR-related disorders and may facilitate the development of targeted therapeutic interventions.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP , Proteína Quinasa C beta , Receptores Nicotínicos , Receptores Nicotínicos/metabolismo , Proteína Quinasa C beta/metabolismo , Humanos , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Animales , Transducción de Señal , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Núcleo Celular/metabolismo
4.
Cell Immunol ; 401-402: 104843, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38905771

RESUMEN

Monocyte migration is an important process in inflammation and atherogenesis. Identification of the key signalling pathways that regulate monocyte migration can provide prospective targets for prophylactic treatments in inflammatory diseases. Previous research showed that the focal adhesion kinase Pyk2, Src kinase and MAP kinases play an important role in MCP-1-induced monocyte migration. In this study, we demonstrate that MCP-1 induces iPLA2 activity, which is regulated by PKCß and affects downstream activation of Rac1 and Pyk2. Rac1 interacts directly with iPLA2 and Pyk2, and plays a crucial role in MCP-1-mediated monocyte migration by modulating downstream Pyk2 and p38 MAPK activation. Furthermore, Rac1 is necessary for cell spreading and F-actin polymerization during monocyte adhesion to fibronectin. Finally, we provide evidence that Rac1 controls the secretion of inflammatory mediator vimentin from MCP-1-stimulated monocytes. Altogether, this study demonstrates that the PKCß/iPLA2/Rac1/Pyk2/p38 MAPK signalling cascade is essential for MCP-1-induced monocyte adhesion and migration.


Asunto(s)
Adhesión Celular , Movimiento Celular , Quimiocina CCL2 , Quinasa 2 de Adhesión Focal , Monocitos , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos , Proteína de Unión al GTP rac1 , Humanos , Monocitos/metabolismo , Monocitos/inmunología , Quimiocina CCL2/metabolismo , Adhesión Celular/fisiología , Proteína de Unión al GTP rac1/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína Quinasa C beta/metabolismo , Actinas/metabolismo
5.
Cardiovasc Diabetol ; 23(1): 202, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867293

RESUMEN

The specific pathophysiological pathways through which diabetes exacerbates myocardial ischemia/reperfusion (I/R) injury remain unclear; however, dysregulation of immune and inflammatory cells, potentially driven by abnormalities in their number and function due to diabetes, may play a significant role. In the present investigation, we simulated myocardial I/R injury by inducing ischemia through ligation of the left anterior descending coronary artery in mice for 40 min, followed by reperfusion for 24 h. Previous studies have indicated that protein kinase Cß (PKCß) is upregulated under hyperglycemic conditions and is implicated in the development of various diabetic complications. The Y4 RNA fragment is identified as the predominant small RNA component present in the extracellular vesicles of cardio sphere-derived cells (CDCs), exhibiting notable anti-inflammatory properties in the contexts of myocardial infarction and cardiac hypertrophy. Our investigation revealed that the administration of Y4 RNA into the ventricular cavity of db/db mice following myocardial I/R injury markedly enhanced cardiac function. Furthermore, Y4 RNA was observed to facilitate M2 macrophage polarization and interleukin-10 secretion through the suppression of PKCß activation. The mechanism by which Y4 RNA affects PKCß by regulating macrophage activation within the inflammatory environment involves the inhibition of ERK1/2 phosphorylation In our study, the role of PKCß in regulating macrophage polarization during myocardial I/R injury was investigated through the use of PKCß knockout mice. Our findings indicate that PKCß plays a crucial role in modulating the inflammatory response associated with macrophage activation in db/db mice experiencing myocardial I/R, with a notable exacerbation of this response observed upon significant upregulation of PKCß expression. In vitro studies further elucidated the protective mechanism by which Y4 RNA modulates the PKCß/ERK1/2 signaling pathway to induce M2 macrophage activation. Overall, our findings suggest that Y4 RNA plays an anti-inflammatory role in diabetic I/R injury, suggesting a novel therapeutic approach for managing myocardial I/R injury in diabetic individuals.


Asunto(s)
Modelos Animales de Enfermedad , Macrófagos , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica , Proteína Quinasa C beta , Transducción de Señal , Animales , Proteína Quinasa C beta/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/genética , Macrófagos/metabolismo , Macrófagos/enzimología , Masculino , Interleucina-10/metabolismo , Interleucina-10/genética , Ratones , Cardiomiopatías Diabéticas/enzimología , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/fisiopatología , Células Cultivadas , Fenotipo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Activación de Macrófagos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Función Ventricular Izquierda , Fosforilación
6.
FEBS Lett ; 598(4): 400-414, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38302840

RESUMEN

The insulin and dopaminergic systems in the brain are associated with schizophrenia and Parkinson's disease with respect to etiology and treatment. The present study investigated the crosstalk between the insulin receptor (IR) and dopamine receptor and found that insulin stimulation selectively inhibits signaling of D3 R in a PKCßII-dependent manner. Upon insulin stimulation, E3 ligase enzyme Mdm2 moves out of the nucleus to ubiquitinate PKCßII. Subsequently, ubiquitinated PKCßII translocates to the cell membrane and interacts with D3 R in a phosphorylation-dependent manner at S229/257, resulting in the attenuation of D3 R signaling and initiating clathrin-mediated endocytosis and downregulation. Considering that both IR and D3 R are closely related to some neuropsychosis, this study could provide new molecular insight into the etiology of the disorder.


Asunto(s)
Dopamina , Insulinas , Proteína Quinasa C beta , Ubiquitinación , Transducción de Señal , Ubiquitina/metabolismo , Insulinas/metabolismo
7.
J Am Heart Assoc ; 13(3): e031028, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38293916

RESUMEN

BACKGROUND: Small conductance calcium-activated potassium (SK) channels are largely responsible for endothelium-dependent coronary arteriolar relaxation. Endothelial SK channels are downregulated by the reduced form of nicotinamide adenine dinucleotide (NADH), which is increased in the setting of diabetes, yet the mechanisms of these changes are unclear. PKC (protein kinase C) is an important mediator of diabetes-induced coronary endothelial dysfunction. Thus, we aimed to determine whether NADH signaling downregulates endothelial SK channel function via PKC. METHODS AND RESULTS: SK channel currents of human coronary artery endothelial cells were measured by whole cell patch clamp method in the presence/absence of NADH, PKC activator phorbol 12-myristate 13-acetate, PKC inhibitors, or endothelial PKCα/PKCß knockdown by using small interfering RNA. Human coronary arteriolar reactivity in response to the selective SK activator NS309 was measured by vessel myography in the presence of NADH and PKCß inhibitor LY333531. NADH (30-300 µmol/L) or PKC activator phorbol 12-myristate 13-acetate (30-300 nmol/L) reduced endothelial SK current density, whereas the selective PKCᵦ inhibitor LY333531 significantly reversed the NADH-induced SK channel inhibition. PKCß small interfering RNA, but not PKCα small interfering RNA, significantly prevented the NADH- and phorbol 12-myristate 13-acetate-induced SK inhibition. Incubation of human coronary artery endothelial cells with NADH significantly increased endothelial PKC activity and PKCß expression and activation. Treating vessels with NADH decreased coronary arteriolar relaxation in response to the selective SK activator NS309, and this inhibitive effect was blocked by coadministration with PKCß inhibitor LY333531. CONCLUSIONS: NADH-induced inhibition of endothelial SK channel function is mediated via PKCß. These findings may provide insight into novel therapeutic strategies to preserve coronary microvascular function in patients with metabolic syndrome and coronary disease.


Asunto(s)
Diabetes Mellitus , Forboles , Humanos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C beta/farmacología , Células Endoteliales/metabolismo , Miristatos/metabolismo , Miristatos/farmacología , NAD/metabolismo , Vasodilatación/fisiología , Diabetes Mellitus/metabolismo , Endotelio Vascular/metabolismo , ARN Interferente Pequeño/metabolismo , Acetatos/metabolismo , Acetatos/farmacología , Forboles/metabolismo , Forboles/farmacología
8.
Eur J Cancer Prev ; 33(1): 45-52, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37505453

RESUMEN

OBJECTIVES: Secreted frizzled-related protein 1 (SFRP1) and protein kinase C-B (PRKCB) contribute to cancer progression and angiogenesis. This study intended to detect SFRP1 and PRKCB expression in non-small-cell lung cancer (NSCLC) patients and analyze its association with clinicopathological features. METHODS: A total of 108 NSCLC patients who underwent surgical resection in our hospital between 2012 and 2017 were retrospectively analyzed. SFRP1 and PRKCB expression was detected using immunohistochemical staining. The relationships between SFRP1 and PRKCB expression and clinicopathological data were analyzed using the chi-square method. Kaplan-Meier analysis was used to investigate survival probability over time. The potential risk of NSCLC morbidity associated with SFRP1 and PRKCB levels was analyzed using univariate and multivariate Cox proportional risk models. RESULTS: SFRP1 and PRKCB expression was negative in 114 and 109 of the 180 NSCLC specimens, respectively. SFRP1 expression was significantly associated with TNM stage ( P  < 0.001) and tumor diameter ( P  < 0.001). PRKCB expression was significantly associated with the TNM stage ( P  < 0.001). The correlation between SFRP1 and PRKCB expression was evident ( P  = 0.023). SFRP1(-) or PRKCB(-) patients shows lower survival rates than SFRP1(+) or PRKCB(+) patients ( P < 0.001). SFRP1(-)/PRKCB(-) patients had the worst prognosis ( P < 0.001). Furthermore, the mortality of SFRP1(-) or PRKCB(-) patients was significantly higher than that of SFRP1(+) or PRKCB(+). CONCLUSION: SFRP1 and PRKCB expression can be used to predict prognosis in patients with NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Pronóstico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/cirugía , Estudios Retrospectivos , Modelos de Riesgos Proporcionales , Biomarcadores de Tumor/metabolismo , Proteínas de la Membrana/genética , Péptidos y Proteínas de Señalización Intercelular , Proteína Quinasa C beta
9.
J Biol Chem ; 299(8): 104917, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37315788

RESUMEN

Although aging is associated with progressive adiposity and a decline in liver function, the underlying molecular mechanisms and metabolic interplay are incompletely understood. Here, we demonstrate that aging induces hepatic protein kinase Cbeta (PKCß) expression, while hepatocyte PKCß deficiency (PKCßHep-/-) in mice significantly attenuates obesity in aged mice fed a high-fat diet. Compared with control PKCßfl/fl mice, PKCßHep-/- mice showed elevated energy expenditure with augmentation of oxygen consumption and carbon dioxide production which was dependent on ß3-adrenergic receptor signaling, thereby favoring negative energy balance. This effect was accompanied by induction of thermogenic genes in brown adipose tissue (BAT) and increased BAT respiratory capacity, as well as a shift to oxidative muscle fiber type with an improved mitochondrial function, thereby enhancing oxidative capacity of thermogenic tissues. Furthermore, in PKCßHep-/- mice, we determined that PKCß overexpression in the liver mitigated elevated expression of thermogenic genes in BAT. In conclusion, our study thus establishes hepatocyte PKCß induction as a critical component of pathophysiological energy metabolism by promoting progressive hepatic and extrahepatic metabolic derangements in energy homeostasis, contributing to late-onset obesity. These findings have potential implications for augmenting thermogenesis as a means of combating aging-induced obesity.


Asunto(s)
Hígado , Obesidad , Proteína Quinasa C beta , Animales , Ratones , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Oxidación-Reducción , Proteína Quinasa C beta/deficiencia , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Regulación Enzimológica de la Expresión Génica , Envejecimiento , Transducción de Señal
10.
J Neurosci Res ; 101(8): 1289-1304, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36967123

RESUMEN

Morphine-induced scratching (MIS) is a common adverse effect associated with the use of morphine as analgesia after surgery. However, the treatment of MIS is less than satisfactory due to its unclear mechanism, which needs to be enunciated. We found that intrathecal (i.t.) injections of morphine significantly enhanced scratching behavior in C57BL/6J male mice as well as increased the expressions of protein kinase C ß (PKCß), phosphorylated p38 mitogen-activated protein kinases (MAPK), and ionized calcium-binding adapter molecule 1 (Iba1) within spinal cord dorsal horn. Conversely, using the kappa opioid receptor antagonist nalbuphine significantly attenuated scratching behavior, reduced PKCß expression and p38 phosphorylation, and decreased spinal dorsal horn microglial activation, while PKCδ and KOR expression elevated. Spinal PKCß silencing mitigated MIS and microglial activation. Still, knockdown of PKCδ reversed the inhibitory effect of nalbuphine on MIS and microglial activation, indicating that PKCδ is indispensable for the antipruritic effects of nalbuphine. In contrast, PKCß is crucial for inducing microglial activation in MIS in male mice. Our findings show a distinct itch cascade of morphine, PKCß/p38MAPK, and microglial activation, but an anti-MIS pathway of nalbuphine, PKCδ/KOR, and neuron activation.


Asunto(s)
Morfina , Nalbufina , Ratones , Masculino , Animales , Morfina/farmacología , Nalbufina/farmacología , Nalbufina/metabolismo , Fosforilación , Microglía/metabolismo , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C beta/farmacología , Ratones Endogámicos C57BL , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Reprod Biol ; 23(2): 100754, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36934663

RESUMEN

We investigated the role of protein kinase c (PKC) -α and -ß during the ovarian follicular dynamics using estrous cycle, gonadotropin-induced ovulation, and antral follicle culture, 4-vinylcyclohexene diepoxide (VCD)-induced premature ovarian failure (POF) in the SD rat models. We found the higher activity of PKC during the proestrus stage along with expression of PKC-α during the estrus and metestrus stages of the estrous cycle while PKC-ß expression was increased during the diestrus, proestrus, and estrus stages. In response to pregnant mare gonadotropin (PMSG)-induced follicular recruitment and ovulation, the phosphorylated (Thr-642) PKC-ß was increased. PKC activity inhibition by hispidin during the proestrus stage resulted in decreased antral follicles and corpus luteum. Treatment with hispidin resulted in the downregulation of granulosa cell (GC) biomarker, follicle stimulating hormone receptor (FSHR) expression in the cultured pre-antral follicle. During the forskolin-induced luteinization of human granulosa cells, the expression level of PKC-α and ß (I and II) was decreased. In the POF condition, the activity of total PKC and the expression levels of PKC-α and ß (I and II) were increased. Immunostaining depicted ubiquitous expression of PKC-α in the ovary during the estrous cycle and POF conditions. Taken together, we conclude the association of PKC-α and -ß (I and II) during ovarian follicular dynamics where the expression level of PKC-α is increased, but the expression level of PKC-ß (I and II) is suppressed in the POF condition in the SD rat model.


Asunto(s)
Insuficiencia Ovárica Primaria , Animales , Femenino , Ratas , Gonadotropinas/farmacología , Proteína Quinasa C beta , Ratas Sprague-Dawley
12.
FEBS Lett ; 597(9): 1300-1316, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36775967

RESUMEN

The plasticity of proximal tubular epithelial cells in response to TGFß contributes to the expression of TWIST1 to drive renal fibrosis. The mechanism of TWIST1 expression is not known. We show that both PI3 kinase and its target mTORC2 increase TGFß-induced TWIST1 expression. TGFß enhances phosphorylation on Ser-660 in the protein kinase C ßII (PKCßII) hydrophobic motif site. Remarkably, phosphorylation-deficient PKCßIIS660A, kinase-dead PKCßII, and PKCßII knockdown blocked TWIST1 expression by TGFß. Inhibition of TWIST1 arrested TGFß-induced tubular cell hypertrophy and the expression of fibronectin, collagen I (α2), and α-smooth muscle actin. By contrast, TWIST1 overexpression induced these pathologies. Interestingly, the inhibition of PKCßII reduced these phenomena, which were countered by the expression of TWIST1. These results provide the first evidence for the involvement of the mTORC2-PKCßII axis in TWIST1 expression to promote tubular cell pathology.


Asunto(s)
Serina-Treonina Quinasas TOR , Factor de Crecimiento Transformador beta , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Proteína Quinasa C beta , Células Epiteliales/metabolismo
13.
J Biol Chem ; 299(2): 102882, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36623731

RESUMEN

Store-operated Ca2+ entry is a ubiquitous mechanism for Ca2+ influx in mammalian cells that regulates a variety of physiological processes. The identification of two forms of Orai1, the predominant store-operated channel, Orai1α and Orai1ß, raises the question whether they differentially regulate cell function. Orai1α is the full-length Orai1, containing 301 amino acids, whereas Orai1ß lacks the N-terminal 63 amino acids. Here, using a combination of biochemistry and imaging combined with the use of human embryonic kidney 293 KO cells, missing the native Orai1, transfected with plasmids encoding for either Orai1α or Orai1ß, we show that Orai1α plays a relevant role in agonist-induced NF-κB transcriptional activity. In contrast, functional Orai1ß is not required for the activation of these transcription factors. The role of Orai1α in the activation of NF-κB is entirely dependent on Ca2+ influx and involves PKCß activation. Our results indicate that Orai1α interacts with PKCß2 by a mechanism involving the Orai1α exclusive AKAP79 association region, which strongly suggests a role for AKAP79 in this process. These findings provide evidence of the role of Orai1α in agonist-induced NF-κB transcriptional activity and reveal functional differences between Orai1 variants.


Asunto(s)
Canales de Calcio , FN-kappa B , Proteína ORAI1 , Humanos , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , FN-kappa B/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Transducción de Señal
14.
Life Sci Alliance ; 6(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36717249

RESUMEN

PKCßII, a conventional PKC family member, plays critical roles in the regulation of a variety of cellular functions. Here, we employed loss-of-function approaches and mutants of PKCßII with altered phosphorylation and protein interaction behaviors to identify the cellular mechanisms underlying the activation of PKCßII. Our results show that 3-phosphoinositide-dependent protein kinase-1 (PDK1)-mediated constitutive phosphorylation of PKCßII at the activation loop (T500) is required for phorbol ester-induced nuclear entry and subsequent Mdm2-mediated ubiquitination of PKCßII, whereas ubiquitination of PKCßII is required for the PDK1-mediated inducible phosphorylation of PKCßII at T500 in the nucleus. After moving out of the nucleus, PKCßII interacts with actin, undergoes inducible mTORC2-mediated phosphorylation at the turn motif (T641), interacts with clathrin, and then translocates to the plasma membrane. This overall cascade of cellular events intertwined with the phosphorylation at critical residues and Mdm2-mediated ubiquitination in the nucleus and along with interactions with actin and clathrin plays roles that encompass the core processes of PKC activation.


Asunto(s)
Actinas , Clatrina , Proteína Quinasa C beta , Proteínas Proto-Oncogénicas c-mdm2 , Actinas/metabolismo , Clatrina/metabolismo , Fosforilación , Proteína Quinasa C beta/metabolismo , Ubiquitinación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
15.
J Biochem Mol Toxicol ; 37(1): e23236, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36239013

RESUMEN

Increasing evidence suggests that disruption of neuron activity contributes to the autistic phenotype. Thus, we aimed in this study to explore the role of protein kinase C beta (PKCß) in the regulation of neuron activity in an autism model. The expression of PKCß in the microarray data of autism animal models was obtained from the Gene Expression Omnibus database. Then, mice with autism-like behavior were prepared in EN2 knockout (-/- ) mice. The interaction between PKCß on fat mass and obesity-associated protein (FTO) as well as between PGC-1α and uncoupling protein 1 (UCP1) were characterized. The effect of FTO on the N6 -methyladenosine (m6A) modification level of proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) was assayed. Following transfection of overexpressed PKCß and/or silenced UCP1, effects of PKCß and UCP1 in autism-like behaviors in EN2-/- mice were analyzed. Results showed that PKCß was downregulated in EN2-/- mouse brain tissues or neurons. PKCß promoted the expression and stability of FTO, which downregulated the m6A modification level of PGC-1α to promote its expression. Moreover, PGC-1α positively targeted the expression of UCP1. PKCß knockdown enhanced sociability and spatial exploration ability, and reduced neuron apoptosis in EN2-/- mouse models of autism, which was reversed by UCP1 overexpression. Collectively, PKCß overexpression leads to activation of the FTO/m6A/PGC-1α/UCP1 axis, thus inhibiting neuron apoptosis and providing neuroprotection in mice with autism-like behavior.


Asunto(s)
Trastorno Autístico , Proteínas de Homeodominio , Proteína Quinasa C beta , Animales , Ratones , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Trastorno Autístico/genética , Proteínas de Homeodominio/genética , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteína Quinasa C beta/metabolismo , Proteína Desacopladora 1/metabolismo , Regulación hacia Arriba
16.
Fetal Pediatr Pathol ; 42(2): 241-252, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36062956

RESUMEN

Background: Ewing sarcoma (ES) can be confirmed by identifying the EWSR1-FLI1 fusion transcript. This study is to investigate whether immunostaining (IHC) of PRKCB-a protein directly regulated by EWSR1-FLI1 is a surrogate maker for diagnosing ES in routine practice. Methods: Microarray gene expression analyses were conducted. RKCB IHC was applied to 69 ES confirmed by morphology and molecular methods, and 41 non-Ewing small round cell tumors. EWSR1 rearrangement, EWSR1-FLI1 fusion or t(11;22)(q24;q12) were identified by fluorescence in situ hybridization, reverse transcriptase polymerase chain reaction, or cytogenetic analysis, respectively. Results: Gene array analyses showed significant overexpression of the PRKCB in ES. PRKCB IHC was positive in 19 cases of ES with EWSR1-FLI1 fusion, 3 cases with cytogenetic 11:22 translocation and 59 cases with EWSR1 rearrangement while negative in only one EWSR1 rearranged case. PRKCB IHC is sensitive (98%) and specific (96%) in detecting EWSR1 rearranged ES. Conclusions: PRKCB is a reliable antibody for diagnosing ES in routine practice.


Asunto(s)
Sarcoma de Ewing , Sarcoma , Humanos , Sarcoma de Ewing/diagnóstico , Sarcoma de Ewing/genética , Inmunohistoquímica , Hibridación Fluorescente in Situ , Proteína EWS de Unión a ARN/genética , Biomarcadores , Proteínas de Fusión Oncogénica/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo
17.
Free Radic Biol Med ; 194: 62-70, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36410585

RESUMEN

Intestinal ischemia reperfusion (I/R) is a common clinical pathological process. We previously reported that pharmacological inhibition of protein kinase C (PKC) ßII with a specific inhibitor attenuated gut I/R injury. However, the endogenous regulatory mechanism of PKCßII inactivation is still unclear. Here, we explored the critical role of caveolin-1 (Cav1) in protecting against intestinal I/R injury by regulating PKCßII inactivation. PKCßII translocated to caveolae and bound with Cav1 after intestinal I/R. Cav1 was highly expressed in the intestine of mice with I/R and IEC-6 cells stimulated with hypoxia/reoxygenation (H/R). Cav1-knockout (KO) mice suffered from worse intestinal injury after I/R than wild-type (WT) mice and showed extremely low survival due to exacerbated systemic inflammatory response syndrome (SIRS) and remote organ (lung and liver) injury. Cav1 deficiency resulted in excessive PKCßII activation and increased oxidative stress and apoptosis after intestinal I/R. Full-length Cav1 scaffolding domain peptide (CSP) suppressed excessive PKCßII activation and protected the gut against oxidative stress and apoptosis due to I/R injury. In summary, Cav1 could regulate PKCßII endogenous inactivation to alleviate intestinal I/R injury. This finding may represent a novel therapeutic strategy for the prevention and treatment of intestinal I/R injury.


Asunto(s)
Caveolina 1 , Daño por Reperfusión , Animales , Ratones , Apoptosis , Caveolina 1/genética , Caveolina 1/metabolismo , Isquemia , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Reperfusión , Daño por Reperfusión/metabolismo
18.
Life Sci ; 312: 121245, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36503900

RESUMEN

AIMS: Conventional members of protein kinase C (PKC) family, including PKCßII, are constitutively phosphorylated on three major motifs and located in the cytosol in a primed state. In response to cellular stimuli, PKCßII is activated through inducible phosphorylation and Mdm2-mediated ubiquitination. In this study, we aimed to identify the activation mechanism of PKCßII, focusing on the signaling cascade that regulate the phosphorylation and ubiquitination. MATERIALS AND METHODS: Loss-of-function approaches and mutants of PDK1/PKCßII that display different regulatory properties were used to identify the cellular components and processes responsible for endocytosis. KEY FINDINGS: Phorbol 12-myristate 13-acetate (PMA)-induced phosphorylation and ubiquitination of PKCßII, which are needed for its translocation to the plasma membrane, required the presence of both Gßγ and 14-3-3ε. Gßγ and 14-3-3ε mediated the constitutive phosphorylation of PKCßII by scaffolding PI3K and PDK1 in the cytosol, which is an inactive but required state for the activation of PKCßII by subsequent signals. In response to PMA treatment, the signaling complex translocated to the nucleus with dissociation of PI3K from it. Thereafter, PDK1 stably interacted with 14-3-3ε and was dephosphorylated; PKCßII interacted with Mdm2 along with Gßγ, leading to its ubiquitination at two lysine residues on its C-tail. Finally, PDK1/14-3-3ε and ubiquitinated PKCßII translocated to the plasma membrane. SIGNIFICANCE: As PKCßII mediates a wide range of cellular functions and plays important roles in the pathogenesis of various diseases, our results will provide clues to understand the pathogenesis of PKCßII-related disorders and facilitate their treatment.


Asunto(s)
Núcleo Celular , Proteínas de Unión al GTP , Proteína Quinasa C beta/metabolismo , Fosforilación , Núcleo Celular/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Acetato de Tetradecanoilforbol/metabolismo , Proteínas de Unión al GTP/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
19.
Eur J Histochem ; 66(4)2022 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-36305269

RESUMEN

Diabetic retinopathy (DR) is a common microvascular complication in patients with diabetes mellitus. DR is caused by chronic hyperglycemia and is characterized by progressive loss of vision because of damage to the retinal microvasculature. In this study, we investigated the regulatory role and clinical significance of the vascular endothelial growth factor (VEGF)/protein kinase C (PKC)/endothelin (ET)/nuclear factor-κB (NF-κB)/intercellular adhesion molecule 1 (ICAM-1) signaling pathway in DR using a rat model. Intraperitoneal injections of the VEGF agonist, streptozotocin (STZ) were used to generate the DR model rats. DR rats treated with the VEGF inhibitor (DR+VEGF inhibitor) were used to study the specific effects of VEGF on DR pathology and the underlying mechanisms. DR and DR+VEGF agonist rats were injected with the PKCß2 inhibitor, GF109203X to determine the therapeutic potential of blocking the VEGF/PKC/ET/NF-κB/ICAM-1 signaling pathway. The body weights and blood glucose levels of the rats in all groups were evaluated at 16 weeks. DR-related retinal histopathology was analyzed by hematoxylin and eosin staining. ELISA assay was used to estimate the PKC activity in the retinal tissues. Western blotting and RT-qPCR assays were used to analyze the expression levels of PKC-ß2, VEGF, ETs, NF-κB, and ICAM-1 in the retinal tissues. Immunohistochemistry was used to analyze VEGF and ICAM-1 expression in the rat retinal tissues. Our results showed that VEGF, ICAM-1, PKCß2, ET, and NF-κB expression levels as well as PKC activity were significantly increased in the retinal tissues of the DR and DR+VEGF agonist rat groups compared to the control and DR+VEGF inhibitor rat groups. DR and DR+VEGF agonist rats showed significantly lower body weight and significantly higher retinal histopathology scores and blood glucose levels compared to the control and DR+VEGF inhibitor group rats. However, treatment of DR and DR+VEGF agonist rats with GF109203X partially alleviated DR pathology by inhibiting the VEGF/ PKC/ET/NF-κB/ICAM-1 signaling pathway. In summary, our data demonstrated that inhibition of the VEGF/ PKC/ET/NF-κB/ICAM-1 signaling pathway significantly alleviated DR-related pathology in the rat model. Therefore, VEGF/PKC/ET/NF-κB/ICAM-1 signaling axis is a promising therapeutic target for DR.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Ratas , Animales , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , FN-kappa B/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Quinasa C beta/metabolismo , Glucemia , Diabetes Mellitus Experimental/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/fisiología
20.
Toxicol Sci ; 190(1): 64-78, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36066426

RESUMEN

Nickel oxide nanoparticles (Nano NiO) lead to pulmonary fibrosis, and the mechanisms are associated with epigenetics. This study aimed to clarify the regulatory relationship among long noncoding RNA HOXA transcript antisense RNA myeloid-specific 1 (HOTAIRM1), DNA methylation and expression of protein kinase C beta (PRKCB), and JNK/c-Jun pathway in Nano NiO-induced pulmonary fibrosis. Therefore, we constructed the rat pulmonary fibrosis model by intratracheal instillation of Nano NiO twice a week for 9 weeks and established the collagen deposition model by treating BEAS-2B cells with Nano NiO for 24 h. Here, the DNA methylation pattern was analyzed by whole-genome bisulfite sequencing in rat fibrotic lung tissues. Then, we integrated mRNA transcriptome data and found 93 DNA methylation genes with transcriptional significance. Meanwhile, the data showed that Nano NiO caused the down-regulation of lncRNA HOTAIRM1, the hypomethylation, and up-regulation of PRKCB2, JNK/c-Jun pathway activation, and collagen deposition (the up-regulated Col-I and α-SMA) both in vivo and in vitro. DNMTs inhibitor 5-AZDC attenuated Nano NiO-induced PRKCB2 expression, JNK/c-Jun pathway activation, and collagen deposition, but overexpression of PRKCB2 aggravated the changes mentioned indicators in Nano NiO-induced BEAS-2B cells. Furthermore, JNK/c-Jun pathway inhibitor (SP600125) alleviated Nano NiO-induced excessive collagen formation. Additionally, overexpression of HOTAIRM1 restrained the PRKCB hypomethylation, the activation of JNK/c-Jun pathway, and collagen formation induced by Nano NiO in BEAS-2B cells. In conclusion, these findings demonstrated that HOTAIRM1 could arrest Nano NiO-induced pulmonary fibrosis by suppressing the PRKCB DNA methylation-mediated JNK/c-Jun pathway.


Asunto(s)
Nanopartículas , Fibrosis Pulmonar , ARN Largo no Codificante , Animales , Ratas , Metilación de ADN/genética , MAP Quinasa Quinasa 4/metabolismo , Nanopartículas/efectos adversos , Nanopartículas/toxicidad , Proteína Quinasa C beta/toxicidad , Proteínas Proto-Oncogénicas c-jun/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , ARN Largo no Codificante/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA