Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.509
Filtrer
1.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38928391

RÉSUMÉ

Oil-Gan is the fruit of the genus Phyllanthus emblica L. The fruits have excellent effects on health care and development values. There are many methods for the management of diabetic nephropathy (DN). However, there is a lack of effective drugs for treating DN throughout the disease course. The primary aim of this study was to examine the protective effects (including analyses of urine and blood, and inflammatory cytokine levels) and mechanisms of the ethyl acetate extract of P. emblica (EPE) on db/db mice, an animal model of diabetic nephropathy; the secondary aim was to examine the expression levels of p- protein kinase Cα (PKCα)/t-PKCα in the kidney and its downregulation of vascular endothelial growth factor (VEGF) and fibrosis gene transforming growth factor-ß1 (TGF-ß1) by Western blot analyses. Eight db/m mice were used as the control group. Forty db/db mice were randomly divided into five groups. Treatments included a vehicle, EPE1, EPE2, EPE3 (at doses of 100, 200, or 400 mg/kg EPE), or the comparative drug aminoguanidine for 8 weeks. After 8 weeks of treatment, the administration of EPE to db/db mice effectively controlled hyperglycemia and hyperinsulinemia by markedly lowering blood glucose, insulin, and glycosylated HbA1c levels. The administration of EPE to db/db mice decreased the levels of BUN and creatinine both in blood and urine and reduced urinary albumin excretion and the albumin creatine ratio (UACR) in urine. Moreover, EPE treatment decreased the blood levels of inflammatory cytokines, including kidney injury molecule-1 (KIM-1), C-reactive protein (CRP), and NLR family pyrin domain containing 3 (NLRP3). Our findings showed that EPE not only had antihyperglycemic effects but also improved renal function in db/db mice. A histological examination of the kidney by immunohistochemistry indicated that EPE can improve kidney function by ameliorating glomerular morphological damage following glomerular injury; alleviating proteinuria by upregulating the expression of nephrin, a biomarker of early glomerular damage; and inhibiting glomerular expansion and tubular fibrosis. Moreover, the administration of EPE to db/db mice increased the expression levels of p- PKCα/t-PKCα but decreased the expression levels of VEGF and renal fibrosis biomarkers (TGF-ß1, collagen IV, p-Smad2, p-Smad3, and Smad4), as shown by Western blot analyses. These results implied that EPE as a supplement has a protective effect against renal dysfunction through the amelioration of insulin resistance as well as the suppression of nephritis and fibrosis in a DN model.


Sujet(s)
Diabète expérimental , Néphropathies diabétiques , Modèles animaux de maladie humaine , Phyllanthus emblica , Extraits de plantes , Animaux , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Souris , Phyllanthus emblica/composition chimique , Mâle , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Acétates/composition chimique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Protein kinase C-alpha/métabolisme , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques
2.
J Nat Prod ; 87(6): 1666-1671, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38840407

RÉSUMÉ

Hypoxia-inducible factor 1 (HIF-1) signaling is upregulated in an oxygen-dependent manner under hypoxic conditions. Activation of HIF-1 signaling increases the expression of HIF-1 target genes involved in cell survival, proliferation, and angiogenesis. Therefore, compounds that activate HIF-1 signaling have therapeutic potential in ischemic diseases. Screening for compounds that activate HIF-1 activity identified a microbial metabolite, teleocidin B-4, a PKC activator. Other PKC activators, such as TPA and 10-Me-Aplog-1, also activated HIF-1 activity. PKC activators induced HIF-1α protein accumulation through PKCα/mTORC activation. These results suggest that PKC activators without tumor-promoting activity have potential as therapeutic agents via HIF-1 target gene activation.


Sujet(s)
Sous-unité alpha du facteur-1 induit par l'hypoxie , Protein kinase C-alpha , Transduction du signal , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Protein kinase C-alpha/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme
3.
CNS Neurosci Ther ; 30(6): e14754, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38884369

RÉSUMÉ

AIMS: Islet cell autoantigen 1 (ICA1) is involved in autoimmune diseases and may affect synaptic plasticity as a neurotransmitter. Databases related to Alzheimer's disease (AD) have shown decreased ICA1 expression in patients with AD. However, the role of ICA1 in AD remains unclear. Here, we report that ICA1 expression is decreased in the brains of patients with AD and an AD mouse model. RESULTS: The ICA1 increased the expression of amyloid precursor protein (APP), disintegrin and metalloprotease 10 (ADAM10), and disintegrin and metalloprotease 17 (ADAM17), but did not affect protein half-life or mRNA levels. Transcriptome sequencing analysis showed that ICA1 regulates the G protein-coupled receptor signaling pathway. The overexpression of ICA1 increased PKCα protein levels and phosphorylation. CONCLUSION: Our results demonstrated that ICA1 shifts APP processing to non-amyloid pathways by regulating the PICK1-PKCα signaling pathway. Thus, this study suggests that ICA1 is a novel target for the treatment of AD.


Sujet(s)
Maladie d'Alzheimer , Précurseur de la protéine bêta-amyloïde , Protein kinase C-alpha , Transduction du signal , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Animaux , Protein kinase C-alpha/métabolisme , Protein kinase C-alpha/génétique , Transduction du signal/physiologie , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Souris , Protéines de transport/métabolisme , Protéines de transport/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Mâle , Souris transgéniques , Femelle , Souris de lignée C57BL , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Encéphale/métabolisme , Protéines du cycle cellulaire
4.
Cell Calcium ; 121: 102894, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38728789

RÉSUMÉ

TRPV2 voltage-insensitive, calcium-permeable ion channels play important roles in cancer progression, immune response, and neuronal development. Despite TRPV2's physiological impact, underlying endogenous proteins mediating TRPV2 responses and affected signaling pathways remain elusive. Using quantitative peroxidase-catalyzed (APEX2) proximity proteomics we uncover dynamic changes in the TRPV2-proximal proteome and identify calcium signaling and cell adhesion factors recruited to the molecular channel neighborhood in response to activation. Quantitative TRPV2 proximity proteomics further revealed activation-induced enrichment of protein clusters with biological functions in neural and cellular projection. We demonstrate a functional connection between TRPV2 and the neural immunoglobulin cell adhesion molecules NCAM and L1CAM. NCAM and L1CAM stimulation robustly induces TRPV2 [Ca2+]I flux in neuronal PC12 cells and this TRPV2-specific [Ca2+]I flux requires activation of the protein kinase PKCα. TRPV2 expression directly impacts neurite lengths that are modulated by NCAM or L1CAM stimulation. Hence, TRPV2's calcium signaling plays a previously undescribed, yet vital role in cell adhesion, and TRPV2 calcium flux and neurite development are intricately linked via NCAM and L1CAM cell adhesion proteins.


Sujet(s)
Calcium , Molécule d'adhérence cellulaire neurale L-1 , Molécules d'adhérence cellulaire neurales , Excroissance neuronale , Protéome , Canaux cationiques TRPV , Animaux , Humains , Rats , Calcium/métabolisme , Signalisation calcique , Adhérence cellulaire , Molécule d'adhérence cellulaire neurale L-1/métabolisme , Molécules d'adhérence cellulaire neurales/métabolisme , Neurites/métabolisme , Cellules PC12 , Protein kinase C-alpha/métabolisme , Protéome/métabolisme , Canaux cationiques TRPV/métabolisme , Antigènes CD56/métabolisme
5.
Pflugers Arch ; 476(7): 1041-1064, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38658400

RÉSUMÉ

Signaling of G protein-activated inwardly rectifying K+ (GIRK) channels is an important mechanism of the parasympathetic regulation of the heart rate and cardiac excitability. GIRK channels are inhibited during stimulation of Gq-coupled receptors (GqPCRs) by depletion of phosphatidyl-4,5-bisphosphate (PIP2) and/or channel phosphorylation by protein kinase C (PKC). The GqPCR-dependent modulation of GIRK currents in terms of specific PKC isoform activation was analyzed in voltage-clamp experiments in rat atrial myocytes and in CHO or HEK 293 cells. By using specific PKC inhibitors, we identified the receptor-activated PKC isoforms that contribute to phenylephrine- and angiotensin-induced GIRK channel inhibition. We demonstrate that the cPKC isoform PKCα significantly contributes to GIRK inhibition during stimulation of wildtype α1B-adrenergic receptors (α1B-ARs). Deletion of the α1B-AR serine residues S396 and S400 results in a preferential regulation of GIRK activity by PKCß. As a novel finding, we report that the AT1-receptor-induced GIRK inhibition depends on the activation of the nPKC isoform PKCε whereas PKCα and PKCß do not mainly participate in the angiotensin-mediated GIRK reduction. Expression of the dominant negative (DN) PKCε prolonged the onset of GIRK inhibition and significantly reduced AT1-R desensitization, indicating that PKCε regulates both GIRK channel activity and the strength of the receptor signal via a negative feedback mechanism. The serine residue S418 represents an important phosphorylation site for PKCε in the GIRK4 subunit. To analyze the functional impact of this PKC phosphorylation site for receptor-specific GIRK channel modulation, we monitored the activity of a phosphorylation-deficient (GIRK4 (S418A)) GIRK4 channel mutant during stimulation of α1B-ARs or AT1-receptors. Mutation of S418 did not impede α1B-AR-mediated GIRK inhibition, suggesting that S418 within the GIRK4 subunit is not subject to PKCα-induced phosphorylation. Furthermore, activation of angiotensin receptors induced pronounced GIRK4 (S418A) channel inhibition, excluding that this phosphorylation site contributes to the AT1-R-induced GIRK reduction. Instead, phosphorylation of S418 has a facilitative effect on GIRK activity that was abolished in the GIRK4 (S418A) mutant. To summarize, the present study shows that the receptor-dependent regulation of atrial GIRK channels is attributed to the GqPCR-specific activation of different PKC isoforms. Receptor-specific activated PKC isoforms target distinct phosphorylation sites within the GIRK4 subunit, resulting in differential regulation of GIRK channel activity with either facilitative or inhibitory effects on GIRK currents.


Sujet(s)
Cricetulus , Canaux potassiques rectifiants entrants couplés aux protéines G , Protéine kinase C , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Animaux , Phosphorylation , Cellules HEK293 , Humains , Rats , Protéine kinase C/métabolisme , Cellules CHO , Récepteurs alpha-1 adrénergiques/métabolisme , Myocytes cardiaques/métabolisme , Mâle , Rat Wistar , Protein kinase C-alpha/métabolisme , Isoenzymes/métabolisme
6.
Int Immunopharmacol ; 132: 111933, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38581988

RÉSUMÉ

Transient receptor potential melastatin 7 (TRPM7) is a cation channel that plays a role in the progression of rheumatoid arthritis (RA), yet its involvement in synovial hyperplasia and inflammation has not been determined. We previously reported that TRPM7 affects the destruction of articular cartilage in RA. Herein, we further confirmed the involvement of TRPM7 in fibroblast-like synoviocyte (FLS) proliferation, metastasis and inflammation. We observed increased TRPM7 expression in FLSs derived from human RA patients. Pharmacological inhibition of TRPM7 protected primary RA-FLSs from proliferation, metastasis and inflammation. Furthermore, we found that TRPM7 contributes to RA-FLS proliferation, metastasis and inflammation by increasing the intracellular Ca2+ concentration. Mechanistically, the PKCα-HuR axis was demonstrated to respond to Ca2+ influx, leading to TRPM7-mediated RA-FLS proliferation, metastasis and inflammation. Moreover, HuR was shown to bind to IL-6 mRNA after nuclear translocation, which could be weakened by TRPM7 channel inhibition. Additionally, adeno-associated virus 9-mediated TRPM7 silencing is highly effective at alleviating synovial hyperplasia and inflammation in adjuvant-induced arthritis rats. In conclusion, our findings unveil a novel regulatory mechanism involved in the pathogenesis of RA and suggest that targeting TRPM7 might be a potential strategy for the prevention and treatment of RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Prolifération cellulaire , Interleukine-6 , Protein kinase C-alpha , Cellules synoviales , Canaux cationiques TRPM , Canaux cationiques TRPM/métabolisme , Canaux cationiques TRPM/génétique , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Animaux , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Humains , Interleukine-6/métabolisme , Interleukine-6/génétique , Protein kinase C-alpha/métabolisme , Protein kinase C-alpha/génétique , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Mâle , Rats , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéine-1 similaire à ELAV/métabolisme , Protéine-1 similaire à ELAV/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Cellules cultivées , Inflammation/métabolisme , Inflammation/anatomopathologie , Rat Sprague-Dawley , Femelle , Transduction du signal
7.
J Exp Clin Cancer Res ; 43(1): 97, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38561833

RÉSUMÉ

BACKGROUND: CAR T cell therapy is a promising approach to improve outcomes and decrease toxicities for patients with cancer. While extraordinary success has been achieved using CAR T cells to treat patients with CD19-positive malignancies, multiple obstacles have so far limited the benefit of CAR T cell therapy for patients with solid tumors. Novel manufacturing and engineering approaches show great promise to enhance CAR T cell function against solid tumors. However, similar to single agent chemotherapy approaches, CAR T cell monotherapy may be unable to achieve high cure rates for patients with difficult to treat solid tumors. Thus, combinatorial drug plus CAR T cell approaches are likely required to achieve widespread clinical success. METHODS: We developed a novel, confocal microscopy based, high-content screen to evaluate 1114 FDA approved drugs for the potential to increase expression of the solid tumor antigen B7-H3 on the surface of osteosarcoma cells. Western blot, RT-qPCR, siRNA knockdown and flow cytometry assays were used to validate screening results and identify mechanisms of drug-induced B7-H3 upregulation. Cytokine and cytotoxicity assays were used to determine if drug pre-treatment enhanced B7-H3-CAR T cell effector function. RESULTS: Fifty-five drugs were identified to increase B7-H3 expression on the surface of LM7 osteosarcoma cells using a novel high-content, high-throughput screen. One drug, ingenol-3-angelate (I3A), increased B7-H3 expression by up to 100%, and was evaluated in downstream experiments. Validation assays confirmed I3A increased B7-H3 expression in a biphasic dose response and cell dependent fashion. Mechanistic studies demonstrated that I3A increased B7-H3 (CD276) mRNA, total protein, and cell surface expression via protein kinase C alpha activation. Functionally, I3A induced B7-H3 expression enhanced B7-H3-CAR T cell function in cytokine production and cytotoxicity assays. CONCLUSIONS: This study demonstrates a novel high-content and high-throughput screen can identify drugs to enhance CAR T cell activity. This and other high-content technologies will pave the way to develop clinical trials implementing rational drug plus CAR T cell combinatorial therapies. Importantly, the technique could also be repurposed for an array of basic and translational research applications where drugs are needed to modulate cell surface protein expression.


Sujet(s)
Tumeurs osseuses , Diterpènes , Ostéosarcome , Humains , Protein kinase C-alpha/métabolisme , Antigènes B7/génétique , Antigènes B7/métabolisme , Ostéosarcome/métabolisme , Tumeurs osseuses/anatomopathologie , Lymphocytes T , Cytokines/métabolisme , Lignée cellulaire tumorale
8.
Arterioscler Thromb Vasc Biol ; 44(6): 1202-1221, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38602101

RÉSUMÉ

BACKGROUND: Hypertension is a major, prevalent risk factor for the development and progression of cerebrovascular disease. Regular exercise has been recommended as an excellent choice for the large population of individuals with mild-to-moderate elevations in blood pressure, but the mechanisms that underlie its vascular-protective and antihypertensive effects remain unknown. Here, we describe a mechanism by which myocyte AKAP150 (A-kinase anchoring protein 150) inhibition induced by exercise training alleviates voltage-dependent L-type Ca2+ channel (CaV1.2) activity and restores cerebral arterial function in hypertension. METHODS: Spontaneously hypertensive rats and newly generated smooth muscle-specific AKAP150 knockin mice were used to assess the role of myocyte AKAP150/CaV1.2 channel in regulating cerebral artery function after exercise intervention. RESULTS: Activation of the AKAP150/PKCα (protein kinase Cα) signaling increased CaV1.2 activity and Ca2+ influx of cerebral arterial myocyte, thus enhancing vascular tone in spontaneously hypertensive rats. Smooth muscle-specific AKAP150 knockin mice were hypertensive with higher CaV1.2 channel activity and increased vascular tone. Furthermore, treatment of Ang II (angiotensin II) resulted in a more pronounced increase in blood pressure in smooth muscle-specific AKAP150 knockin mice. Exercise training significantly reduced arterial myocyte AKAP150 expression and alleviated CaV1.2 channel activity, thus restoring cerebral arterial function in spontaneously hypertensive rats and smooth muscle-specific AKAP150 knockin mice. AT1R (AT1 receptor) and AKAP150 were interacted closely in arterial myocytes. Exercise decreased the circulating Ang II and Ang II-involved AT1R-AKAP150 association in myocytes of hypertension. CONCLUSIONS: The current study demonstrates that aerobic exercise ameliorates CaV1.2 channel function via inhibiting myocyte AKAP150, which contributes to reduced cerebral arterial tone in hypertension.


Sujet(s)
Protéines d'ancrage aux protéines kinases A , Canaux calciques de type L , Artères cérébrales , Modèles animaux de maladie humaine , Hypertension artérielle , Muscles lisses vasculaires , Myocytes du muscle lisse , Rats de lignée SHR , Animaux , Protéines d'ancrage aux protéines kinases A/métabolisme , Protéines d'ancrage aux protéines kinases A/génétique , Canaux calciques de type L/métabolisme , Canaux calciques de type L/génétique , Hypertension artérielle/physiopathologie , Hypertension artérielle/métabolisme , Hypertension artérielle/génétique , Artères cérébrales/métabolisme , Artères cérébrales/physiopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/physiopathologie , Mâle , Myocytes du muscle lisse/métabolisme , Conditionnement physique d'animal/physiologie , Protein kinase C-alpha/métabolisme , Protein kinase C-alpha/génétique , Signalisation calcique , Souris de lignée C57BL , Souris , Rats , Rats de lignée WKY , Angiotensine-II , Pression sanguine , Transduction du signal
9.
Int J Mol Sci ; 25(5)2024 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-38474189

RÉSUMÉ

Coronary artery spasm (CAS) plays an important role in the pathogeneses of various ischemic heart diseases and has gradually become a common cause of life-threatening arrhythmia. The specific molecular mechanism of CAS has not been fully elucidated, nor are there any specific diagnostic markers for the condition. Therefore, this study aimed to examine the specific molecular mechanism underlying CAS, and screen for potential diagnostic markers. To this end, we successfully constructed a rat CAS model and achieved in vitro culture of a human coronary-artery smooth-muscle cell (hCASMC) contraction model. Possible molecular mechanisms by which protein kinase C (PKC) regulated CAS through the C kinase-potentiated protein phosphatase 1 inhibitor of 17 kDa (CPI-17)/myosin II regulatory light chain (MLC2) pathway were studied in vivo and in vitro to screen for potential molecular markers of CAS. We performed hematoxylin and eosin staining, myocardial zymogram, and transmission electron microscopy to determine myocardial and coronary artery injury in CAS rats. Then, using immunohistochemical staining, immunofluorescence staining, and Western blotting, we further demonstrated a potential molecular mechanism by which PKC regulated CAS via the CPI-17/MLC2 pathway. The results showed that membrane translocation of PKCα occurred in the coronary arteries of CAS rats. CPI-17/MLC2 signaling was observably activated in coronary arteries undergoing CAS. In addition, in vitro treatment of hCASMCs with angiotensin II (Ang II) increased PKCα membrane translocation while consistently activating CPI-17/MLC2 signaling. Conversely, GF-109203X and calphostin C, specific inhibitors of PKC, inactivated CPI-17/MLC2 signaling. We also collected the coronary artery tissues from deceased subjects suspected to have died of CAS and measured their levels of phosphorylated CPI-17 (p-CPI-17) and MLC2 (p-MLC2). Immunohistochemical staining was positive for p-CPI-17 and p-MLC2 in the tissues of these subjects. These findings suggest that PKCα induced CAS through the CPI-17/MLC2 pathway; therefore, p-CPI-17 and p-MLC2 could be used as potential markers for CAS. Our data provide novel evidence that therapeutic strategies against PKC or CPI-17/MLC2 signaling might be promising in the treatment of CAS.


Sujet(s)
Spasme coronaire , Animaux , Humains , Rats , Marqueurs biologiques/métabolisme , Mort subite cardiaque , Phosphoprotéines/métabolisme , Phosphorylation , Protéine kinase C/métabolisme , Protein kinase C-alpha/métabolisme
10.
Food Funct ; 15(8): 4180-4192, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38506030

RÉSUMÉ

Until now, the beneficial vascular properties of Hop reported in the literature have been mainly attributed to specific compound classes, such as tannins and phenolic acids. However, the potential vascular action of a Hop subfraction containing a high amount of α or ß acids remains completely understood. Therefore, this study aims to investigate the vascular effects of the entire Hop extract and to fraction the Hop extract to identify the main bioactive vascular compounds. A pressure myograph was used to perform vascular reactivity studies on mouse resistance arteries. Phytocomplex fractionation was performed on a semi-prep HPLC system and characterized by UHPLC-PDA-MS/MS coupled to mass spectrometry. Western blot analysis was performed to characterize the phosphorylation site enrolled. The entire Hop extract exerts a direct dose-dependent endothelial vascular action. The B1 subfraction, containing a high concentration of α acids, recapitulates the vascular effect of the crude extract. Its vasorelaxant action is mediated by the opening of Transient Receptor Potential Vanilloid type 4 (TRPV4), potentiated by PKCα, and subsequent involvement of endothelial small-conductance calcium-activated potassium channels (SKCa) and intermediate-conductance calcium-activated potassium channels (IKCa) that drives endothelium-dependent hyperpolarization (EDH) through heterocellular myoendothelial gap junctions (MEGJs). This is the first comprehensive investigation of the vascular function of Hop-derived α acids in resistance arteries. Overall, our data suggest that the B1 subfraction from Hop extracts, containing only α acids, has great potential to be translated into the useful armamentarium of natural bioactive compounds with cardiovascular benefits.


Sujet(s)
Humulus , Extraits de plantes , Protein kinase C-alpha , Canaux cationiques TRPV , Vasodilatateurs , Humulus/composition chimique , Animaux , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Protein kinase C-alpha/métabolisme , Canaux cationiques TRPV/métabolisme , Souris , Vasodilatateurs/pharmacologie , Vasodilatateurs/composition chimique , Mâle , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Humains , Vasodilatation/effets des médicaments et des substances chimiques , Souris de lignée C57BL
11.
Am J Pathol ; 194(5): 796-809, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38395146

RÉSUMÉ

α-Synuclein (α-Syn) is a key determinator of Parkinson disease (PD) pathology, but synapse and microcircuit pathologies in the retina underlying visual dysfunction are poorly understood. Herein, histochemical and ultrastructural analyses and ophthalmologic measurements in old transgenic M83 PD model (mice aged 16 to 18 months) indicated that abnormal α-Syn aggregation in the outer plexiform layer (OPL) was associated with degeneration in the C-terminal binding protein 2 (CtBP2)+ ribbon synapses of photoreceptor terminals and protein kinase C alpha (PKCα)+ rod bipolar cell terminals, whereas α-Syn aggregates in the inner retina correlated with the reduction and degeneration of tyrosine hydroxylase- and parvalbumin-positive amacrine cells. Phosphorylated Ser129 α-synuclein expression was strikingly restricted in the OPL, with the most severe degenerations in the entire retina, including mitochondrial degeneration and loss of ribbon synapses in 16- to 18-month-old mice. These synapse- and microcircuit-specific deficits of the rod pathway at the CtBP2+ rod terminals and PKCα+ rod bipolar and amacrine cells were associated with attenuated a- and b-wave amplitudes and oscillatory potentials on the electroretinogram. They were also associated with the impairment of visual functions, including reduced contrast sensitivity and impairment of the middle range of spatial frequencies. Collectively, these findings demonstrate that α-Syn aggregates cause the synapse- and microcircuit-specific deficits of the rod pathway and the most severe damage to the OPL, providing the retinal synaptic and microcircuit basis for visual dysfunctions in PD.


Sujet(s)
Protein kinase C-alpha , alpha-Synucléine , Animaux , Souris , alpha-Synucléine/métabolisme , Cellules amacrines/métabolisme , Protein kinase C-alpha/métabolisme , Rétine/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/ultrastructure , Synapses/métabolisme , Facteurs de transcription/métabolisme
12.
Mol Biotechnol ; 66(5): 1062-1070, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38184808

RÉSUMÉ

To investigate the inhibitory effect of hirudin on the cell proliferation of human ovarian cancer A2780 cells by preventing thrombin and its underlying molecular mechanism. Cell Counting Kit-8 (CCK-8) method was used to detect the effect of different concentrations of hirudin and thrombin on the cell proliferation of A2780 cells. PAR-1 wild-type overexpression plasmid was constructed utilizing enzyme digestion identification, and it was transferred to A2780 cells. Sequencing and Western blot were used to detect the changes in PAR-1 protein expression. Western blot detection of PKCα protein phosphorylation in A2780 cells was performed. We also implemented quantitative PCR to detect the mRNA expression levels of epithelial-mesenchymal transition (EMT)-related genes, CDH2, Snail, and Vimentin, in A2780 cells. 1 µg/ml hirudin treatment maximally inhibited the promotion of A2780 cell proliferation by thrombin. Hirudin inhibited the binding of thrombin to the N-terminus of PAR-1, hindered PKCα protein phosphorylation in A2780 cells, and downregulated the mRNA expression levels of CDH2, Snail, and Vimentin. In conclusion, hirudin inhibits the cell proliferation of ovarian cancer A2780 cells, and the underlying mechanism may be through downregulating the transcription level of EMT genes, CDH2, Snail, and Vimentin. This study indicates that hirudin may have a therapeutic potential as an anti-cancer agent for ovarian cancer.


Sujet(s)
Prolifération cellulaire , Transition épithélio-mésenchymateuse , Hirudines , Tumeurs de l'ovaire , Humains , Hirudines/pharmacologie , Hirudines/génétique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Protein kinase C-alpha/métabolisme , Protein kinase C-alpha/génétique , Récepteur de type PAR-1/génétique , Récepteur de type PAR-1/métabolisme , Thrombine/pharmacologie , Thrombine/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Vimentine/métabolisme , Vimentine/génétique
13.
J Cell Mol Med ; 28(3): e18110, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38164042

RÉSUMÉ

BACKGROUND AND AIMS: The secretion of bile salts transported by the bile salt export pump (BSEP) is the primary driving force for the generation of bile flow; thus, it is closely related to the formation of cholesterol stones. Caveolin-1 (Cav-1), an essential player in cell signalling and endocytosis, is known to co-localize with cholesterol-rich membrane domains. This study illustrates the role of Cav-1 and BSEP in cholesterol stone formation. METHODS: Adult male C57BL/6 mice were used as an animal model. HepG2 cells were cultured under different cholesterol concentrations and BSEP, Cav-1, p-PKCα and Hax-1 expression levels were determined via Western blotting. Expression levels of BSEP and Cav-1 mRNA were detected using real-time PCR. Immunofluorescence and immunoprecipitation assays were performed to study BSEP and Hax-1 distribution. Finally, an ATPase activity assay was performed to detect BSEP transport activity under different cholesterol concentrations in cells. RESULTS: Under low-concentration stimulation with cholesterol, Cav-1 and BSEP protein and mRNA expression levels significantly increased, PKCα phosphorylation significantly decreased, BSEP binding capacity to Hax-1 weakened, and BSEP function increased. Under high-concentration stimulation with cholesterol, Cav-1 and BSEP protein and mRNA expression levels decreased, PKCα phosphorylation increased, BSEP binding capacity to Hax-1 rose, and BSEP function decreased. CONCLUSION: Cav-1 regulates the bile salt export pump on the canalicular membrane of hepatocytes via PKCα-associated signalling under cholesterol stimulation.


Sujet(s)
Cavéoline-1 , Protein kinase C-alpha , Animaux , Mâle , Souris , Membre-11 de la sous-famille B à cassette liant l'ATP/métabolisme , Transporteurs ABC/génétique , Acides et sels biliaires/métabolisme , Cavéoline-1/métabolisme , Cholestérol/métabolisme , Hépatocytes/métabolisme , Souris de lignée C57BL , Protein kinase C-alpha/métabolisme , ARN messager/métabolisme , Humains
14.
Cancer Res ; 84(4): 560-576, 2024 02 15.
Article de Anglais | MEDLINE | ID: mdl-38117512

RÉSUMÉ

Phospholipase C epsilon 1 (PLCE1) is a well-established susceptibility gene for esophageal squamous cell carcinoma (ESCC). Identification of the underlying mechanism(s) regulated by PLCE1 could lead to a better understanding of ESCC tumorigenesis. In this study, we found that PLCE1 enhances tumor progression by regulating the replicative helicase MCM7 via two pathways. PLCE1 activated PKCα-mediated phosphorylation of E2F1, which led to the transcriptional activation of MCM7 and miR-106b-5p. The increased expression of miR-106b-5p, located in intron 13 of MCM7, suppressed autophagy and apoptosis by targeting Beclin-1 and RBL2, respectively. Moreover, MCM7 cooperated with the miR-106b-25 cluster to promote PLCE1-dependent cell-cycle progression both in vivo and in vitro. In addition, PLCE1 potentiated the phosphorylation of MCM7 at six threonine residues by the atypical kinase RIOK2, which promoted MCM complex assembly, chromatin loading, and cell-cycle progression. Inhibition of PLCE1 or RIOK2 hampered MCM7-mediated DNA replication, resulting in G1-S arrest. Furthermore, MCM7 overexpression in ESCC correlated with poor patient survival. Overall, these findings provide insights into the role of PLCE1 as an oncogenic regulator, a promising prognostic biomarker, and a potential therapeutic target in ESCC. SIGNIFICANCE: PLCE1 promotes tumor progression in ESCC by activating PKCα-mediated phosphorylation of E2F1 to upregulate MCM7 and miR-106b-5p expression and by potentiating MCM7 phosphorylation by RIOK2.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , microARN , Humains , Tumeurs de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/métabolisme , Phosphorylation , Protein kinase C-alpha/métabolisme , Lignée cellulaire tumorale , Phosphoinositide Phospholipase C/génétique , Phosphoinositide Phospholipase C/métabolisme , microARN/génétique , microARN/métabolisme , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire , Composant-7 du complexe de maintenance des minichromosomes/génétique , Composant-7 du complexe de maintenance des minichromosomes/métabolisme
15.
Food Funct ; 15(2): 779-793, 2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38126185

RÉSUMÉ

As a common plant-derived dietary flavonoid, rutin receives widespread attention because of its good antioxidant bioactivities. Protein kinase Cα (PKCα) is a serine/threonine kinase that is involved in uncountable cellular processes, among which ferroptosis, a novel form of cell death, is triggered by lipid peroxidation and has been reported to be associated with pulmonary arterial hypertension (PAH). But it is still not well appreciated how rutin inhibits ferroptosis in PAH and what function PKCα has in this process. In this study, we first observed whether rutin could prevent PAH by attenuating ferroptosis with a PAH animal model and pulmonary artery smooth muscle cells (PASMCs) under hypoxia. Mitochondrial metabolomics and network pharmacology were employed to clarify the metabolic alterations and screen target proteins, and the results showed that PKCα was a vital node in rutin regulating mitochondrial metabolism related to ferroptosis in PAH. Based on molecular docking and multispectral analysis, we found that rutin could directly interact with PKCα through hydrogen bonds, which could induce static quenching, and then influence the secondary structure of PKCα. In conclusion, these findings mainly point to a novel mechanism that rutin protects PAH rats by modifying the structure and altering the activity of PKCα, and thus suppressing ferroptosis. This work reveals that the interaction behaviors between small molecules and bio-macromolecules are a critical factor to develop natural biological active ingredients and gives an insight into the potential applications of flavonoids in health and disease.


Sujet(s)
Ferroptose , Hypertension pulmonaire , Hypertension artérielle pulmonaire , Rats , Animaux , Hypertension artérielle pulmonaire/complications , Hypertension artérielle pulmonaire/métabolisme , Hypertension pulmonaire/traitement médicamenteux , Hypertension pulmonaire/étiologie , Rutoside/métabolisme , Protein kinase C-alpha/génétique , Protein kinase C-alpha/métabolisme , Simulation de docking moléculaire , Prolifération cellulaire , Myocytes du muscle lisse/métabolisme , Cellules cultivées
16.
Mol Biol Cell ; 35(1): ar5, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37910189

RÉSUMÉ

The interaction between aggregated low-density lipoprotein (agLDL) and macrophages in arteries plays a major role in atherosclerosis. Macrophages digest agLDL and generate free cholesterol in an extracellular, acidic, hydrolytic compartment known as the lysosomal synapse. Macrophages form a tight seal around agLDL through actin polymerization and deliver lysosomal contents into this space in a process termed digestive exophagy. Our laboratory has identified TLR4 activation of MyD88/Syk as critical for digestive exophagy. Here we use pharmacological agents and siRNA knockdown to characterize signaling pathways downstream of Syk that are involved in digestive exophagy. Syk activates Bruton's tyrosine kinase (BTK) and phospholipase Cγ2 (PLCγ2). We show that PLCγ2 and to a lesser extent BTK regulate digestive exophagy. PLCγ2 cleaves PI(4,5)P2 into diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). Soluble IP3 activates release of Ca2+ from the endoplasmic reticulum (ER). We demonstrate that Ca2+ release from the ER is upregulated by agLDL and plays a key role in digestive exophagy. Both DAG and Ca2+ activate protein kinase Cα (PKCα). We find that PKCα is an important regulator of digestive exophagy. These results expand our understanding of the mechanisms of digestive exophagy, which could be useful in developing therapeutic interventions to slow development of atherosclerosis.


Sujet(s)
Athérosclérose , Protein kinase C-alpha , Humains , Protein kinase C-alpha/métabolisme , Phospholipase C gamma/métabolisme , Macrophages/métabolisme , Transduction du signal , Lipoprotéines LDL/métabolisme , Agammaglobulinaemia tyrosine kinase/métabolisme , Athérosclérose/métabolisme , Digestion
17.
Exp Biol Med (Maywood) ; 248(23): 2440-2448, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38158699

RÉSUMÉ

The mammalian target of rapamycin (mTOR) inhibitors, everolimus (but not dactolisib), is frequently associated with lung injury in clinical therapies. However, the underlying mechanisms remain unclear. Endothelial cell barrier dysfunction plays a major role in the pathogenesis of the lung injury. This study hypothesizes that everolimus increases pulmonary endothelial permeability, which leads to lung injury. We tested the effects of everolimus on human pulmonary microvascular endothelial cell (HPMEC) permeability and a mouse model of intraperitoneal injection of everolimus was established to investigate the effect of everolimus on pulmonary vascular permeability. Our data showed that everolimus increased human pulmonary microvascular endothelial cell (HPMEC) permeability which was associated with MLC phosphorylation and F-actin stress fiber formation. Furthermore, everolimus induced an increasing concentration of intracellular calcium Ca2+ leakage in HPMECs and this was normalized with ryanodine pretreatment. In addition, ryanodine decreased everolimus-induced phosphorylation of PKCα and MLC, and barrier disruption in HPMECs. Consistent with in vitro data, everolimus treatment caused a visible lung-vascular barrier dysfunction, including an increase in protein in BALF and lung capillary-endothelial permeability, which was significantly attenuated by pretreatment with an inhibitor of PKCα, MLCK, and ryanodine. This study shows that everolimus induced pulmonary endothelial hyper-permeability, at least partly, in an MLC phosphorylation-mediated EC contraction which is influenced in a Ca2+-dependent manner and can lead to lung injury through mTOR-independent mechanisms.


Sujet(s)
Cellules endothéliales , Lésion pulmonaire , Animaux , Souris , Humains , Cellules endothéliales/métabolisme , Évérolimus/pharmacologie , Évérolimus/métabolisme , Lésion pulmonaire/anatomopathologie , Endothélium vasculaire , Protein kinase C-alpha/métabolisme , Protein kinase C-alpha/pharmacologie , Ryanodine/métabolisme , Ryanodine/pharmacologie , Poumon/métabolisme , Phosphorylation , Cellules cultivées , Sérine-thréonine kinases TOR/métabolisme , Mammifères
18.
Sci Rep ; 13(1): 22558, 2023 12 18.
Article de Anglais | MEDLINE | ID: mdl-38110453

RÉSUMÉ

Diabetes mellitus (DM) is a common chronic metabolic disease in humans and household cats that is characterized by persistent hyperglycemia. DM is associated with dysfunction of the intestinal barrier. This barrier is comprised of an epithelial monolayer that contains a network of tight junctions that adjoin cells and regulate paracellular movement of water and solutes. The mechanisms driving DM-associated barrier dysfunction are multifaceted, and the direct effects of hyperglycemia on the epithelium are poorly understood. Preliminary data suggest that fenofibrate, An FDA-approved peroxisome proliferator-activated receptor-alpha (PPARα) agonist drug attenuates intestinal barrier dysfunction in dogs with experimentally-induced DM. We investigated the effects of hyperglycemia-like conditions and fenofibrate treatment on epithelial barrier function using feline intestinal organoids. We hypothesized that glucose treatment directly increases barrier permeability and alters tight junction morphology, and that fenofibrate administration can ameliorate these deleterious effects. We show that hyperglycemia-like conditions directly increase intestinal epithelial permeability, which is mitigated by fenofibrate. Moreover, increased permeability is caused by disruption of tight junctions, as evident by increased junctional tortuosity. Finally, we found that increased junctional tortuosity and barrier permeability in hyperglycemic conditions were associated with increased protein kinase C-α (PKCα) activity, and that fenofibrate treatment restored PKCα activity to baseline levels. We conclude that hyperglycemia directly induces barrier dysfunction by disrupting tight junction structure, a process that is mitigated by fenofibrate. We further propose that counteracting modulation of PKCα activation by increased intracellular glucose levels and fenofibrate is a key candidate regulatory pathway of tight junction structure and epithelial permeability.


Sujet(s)
Fénofibrate , Hyperglycémie , Maladies intestinales , Humains , Chats , Animaux , Chiens , Glucose/pharmacologie , Glucose/métabolisme , Protein kinase C-alpha/métabolisme , Fénofibrate/pharmacologie , Intestins , Hyperglycémie/métabolisme , Maladies intestinales/métabolisme , Jonctions serrées/métabolisme , Muqueuse intestinale/métabolisme , Perméabilité
19.
Biomed Pharmacother ; 169: 115917, 2023 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38006617

RÉSUMÉ

BACKGROUND: Glutamate stimuli and hyperactivation of its receptor are predominant determinants of ischemia-induced cytotoxic cerebral edema, which is closely associated with protein nanoparticle (PN)-induced increases in osmotic pressure. Herein, we investigated the electrochemical and mechanical mechanisms underlying the neuron swelling induced by PNs via the co-activation of N-methyl-D-aspartate receptor subunit (NMDAR) and excitatory metabotropic glutamate receptors (mGluRs). RESULTS: We observed that co-activation of ionic glutamate receptor NMDAR and Group I metabotropic mGluRs promoted alteration of PN-induced membrane potential and increased intracellular osmosis, which was closely associated with calcium and voltage-dependent ion channels. In addition, activation of NMDAR-induced calmodulin (CaM) and mGluR downstream diacylglycerol (DAG)/protein kinase C α (PKCα) were observed to play crucial roles in cytotoxic hyperosmosis. The crosstalk between CaM and PKCα could upregulate the sensitivity and sustained opening of sulfonylurea receptor 1 (SUR1)-transient receptor potential cation channel subfamily M member 4 (TRPM4) and transmembrane protein 16 A (TMEM16A) channels, respectively, maintaining the massive Na+/Cl- influx, and the resultant neuron hyperosmosis and swelling. Intracellular PNs and Na+/Cl- influx were found to be as potential targets for cerebral edema treatment, using the neurocyte osmosis system and a cerebral ischemic rat model. CONCLUSIONS: This study highlights PNs as a key factor in "electrochemistry-tension" signal transduction controlling Na+/Cl- ion channels and increased osmotic pressure in ischemia-induced cytotoxic edema. Moreover, enhanced sensitivity in both Na+ and Cl- ion channels also has a crucial role in cerebral edema.


Sujet(s)
Oedème cérébral , Nanoparticules , Récepteurs métabotropes au glutamate , Canaux cationiques TRPM , Rats , Animaux , Récepteurs du N-méthyl-D-aspartate/physiologie , Récepteurs métabotropes au glutamate/métabolisme , Pression osmotique , Protein kinase C-alpha/métabolisme , Oedème , Ischémie , Canaux cationiques TRPM/métabolisme
20.
Int J Mol Sci ; 24(19)2023 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-37833954

RÉSUMÉ

Acute hepatitis (AH) is a common liver disease with an increasing number of patients each year, requiring the development of new treatments. Hence, our work aimed to evaluate the therapeutic effect of Oryza sativa L. indica (purple rice) seed coat on concanavalin A (ConA)-induced AH and further reveal its potential mechanisms. Purple rice seed coat extract (PRE) was extracted with hydrochloric acid ethanol and analyzed through a widely targeted components method. We evaluated the effects of PRE on AH through histopathological examination, liver function, gut microbiota composition, and the intestinal barrier. The potential targets of PRE on AH were predicted by bioinformatics. Western blotting, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) staining, and corresponding kits were used to investigate PRE effects on predicting targets and associated signaling pathways in AH mice. In AH model mice, PRE treatment increased transformed mouse 3T3 cell double minute 2 (MDM2) expression to inhibit apoptosis; it also markedly downregulated protein kinase C alpha (PKCα), prostaglandin-endoperoxide synthase 1 (PTGS1), and mitogen-activated protein kinase 1 (MAPK1) activity to alleviate inflammation. Thus, PRE treatment also recovered the intestinal barrier, decreased the lipopolysaccharide (LPS) levels of plasma and the liver, enhanced liver function, and improved the composition of intestinal microbiota. In general, PRE targeting MDM2, PKCα, MAPK1, and PTGS1 ameliorated ConA-induced AH by attenuating inflammation and apoptosis, restoring the intestinal barrier, enhancing the liver function, and improving the gut microbiota, which revealed that the purple rice seed coat might hold possibilities as a therapeutic option for AH.


Sujet(s)
Hépatite , Oryza , Humains , Animaux , Souris , Oryza/métabolisme , Concanavaline A/toxicité , Concanavaline A/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Mitogen-Activated Protein Kinase 1/métabolisme , Protein kinase C-alpha/métabolisme , Hépatite/traitement médicamenteux , Hépatite/étiologie , Hépatite/métabolisme , Transduction du signal , Maladie aigüe , Inflammation , Protéines proto-oncogènes c-mdm2/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...