Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Development ; 151(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38078543

RESUMO

The kinase PAR-4/LKB1 is a major regulator of intestinal homeostasis, which prevents polyposis in humans. Moreover, its ectopic activation is sufficient to induce polarization and formation of microvilli-like structures in intestinal cell lines. Here, we use Caenorhabditis elegans to examine the role of PAR-4 during intestinal development in vivo. We show that it is not required to establish enterocyte polarity and plays only a minor role in brush border formation. By contrast, par-4 mutants display severe deformations of the intestinal lumen as well as supernumerary intestinal cells, thereby revealing a previously unappreciated function of PAR-4 in preventing intestinal hyperplasia. The presence of supernumerary enterocytes in par-4 mutants is not due to excessive cell proliferation, but rather to the abnormal expression of the intestinal cell fate factors end-1 and elt-2 outside the E lineage. Notably, par-4 mutants also display reduced expression of end-1 and elt-2 inside the E lineage. Our work thereby unveils an essential and dual role of PAR-4, which both restricts intestinal specification to the E lineage and ensures its robust differentiation.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Humanos , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/metabolismo , Polaridade Celular , Endoderma/metabolismo , Hiperplasia/metabolismo , Intestinos , Embrião não Mamífero/metabolismo
2.
Am J Physiol Renal Physiol ; 326(2): F219-F226, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38031732

RESUMO

Protease-activated receptor 4 (PAR4) is a G protein-coupled receptor activated by thrombin. In the platelet, response to thrombin PAR4 contributes to the predominant procoagulant microparticle formation, increased fibrin deposition, and initiation of platelet-stimulated inflammation. In addition, PAR4 is expressed in other cell types, including endothelial cells. Under inflammatory conditions, PAR4 is overexpressed via epigenetic demethylation of the PAR4 gene, F2RL3. PAR4 knockout (KO) studies have determined a role for PAR4 in ischemia-reperfusion injury in the brain, and PAR4 KO mice display normal cardiac function but present less myocyte death and cardiac dysfunction in response to acute myocardial infarction. Although PAR4 has been reported to be expressed within the kidney, the contribution of PAR4 to acute kidney injury (AKI) and chronic kidney disease (CKD) is not well understood. Here we report that PAR4 KO mice are protected against kidney injury in two mouse models. First, PAR4 KO mice are protected against induction of markers of both fibrosis and inflammation in two different models of kidney injury: 1) 7 days following unilateral ureter obstruction (UUO) and 2) an AKI-CKD model of ischemia-reperfusion followed by 8 days of contralateral nephrectomy. We further show that PAR4 expression in the kidney is low in the control mouse kidney but induced over time following UUO. PAR4 KO mice are protected against blood urea nitrogen (BUN) and glomerular filtration rate (GFR) kidney function pathologies in the AKI-CKD model. Following the AKI-CKD model, PAR4 is expressed in the collecting duct colocalizing with Dolichos biflorus agglutinin (DBA), but not in the proximal tubule with Lotus tetragonolobus lectin (LTL). Collectively, the results reported in this study implicate PAR4 as contributing to the pathology in mouse models of acute and chronic kidney injury.NEW & NOTEWORTHY The contribution of the thrombin receptor protease-activated receptor 4 (PAR4) to acute kidney injury (AKI) and chronic kidney disease (CKD) is not well understood. Here we report that PAR4 expression is upregulated after kidney injury and PAR4 knockout (KO) mice are protected against fibrosis following kidney injury in two mouse models. First, PAR4 KO mice are protected against unilateral ureter obstruction. Second, PAR4 KO mice are protected against an AKI-CKD model of ischemia-reperfusion followed by contralateral nephrectomy.


Assuntos
Injúria Renal Aguda , Insuficiência Renal Crônica , Animais , Camundongos , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Células Endoteliais/metabolismo , Fibrose , Inflamação/patologia , Isquemia/patologia , Rim/metabolismo , Camundongos Knockout , Receptores de Trombina/genética , Receptores de Trombina/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Traumatismo por Reperfusão/patologia , Trombina/metabolismo , Trombina/farmacologia
3.
Ann Hepatol ; 29(6): 101538, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39147129

RESUMO

INTRODUCTION AND OBJECTIVES: Prostate apoptosis response protein-4 (PAR-4) is considered a tumor suppressor. However, the role of PAR-4 in hepatocellular carcinoma (HCC) has rarely been reported. The study explores the role of PAR-4 in the malignant behaviors of HCC cells. MATERIALS AND METHODS: TCGA database was applied to analyze the expression of PAR-4 in HCC. Evaluated PAR-4 relationship with clinical parameters and prognosis by tissue microarray; expression of STAT3, p-STAT3, Src and Ras was detected by Western blotting or laser confocal microscopy. Cell scratch and flow cytometry assays were used to observe IL-6 regulation of the malignant behaviors of HCC cells. The tumorigenic potential of HCC cells in vivo was evaluated in a nude mouse tumor model. RESULTS: Analysis indicated that the expression of PAR-4 in HCC tissues was significantly higher than that in normal liver tissues; and PAR-4 interacted with STAT3. KEGG analysis showed that PAR-4 plays a role in the Janus kinase (JAK)/STAT signaling pathway. The positive expression rate of PAR-4 in HCC tissues was significantly higher than that in adjacent tissues. Positive correlation between IL-6 and PAR-4 expression in the HCC tissues. Exogenous IL-6 significantly promoted the proliferation and migration of HCC cells and up-regulated the expression of PAR-4 and p-STAT3 in HCC cells. Interference of the expression of PAR-4 could reduce the malignant behaviors of HCC cells and inhibit tumorigenesis in a nude mouse tumor model. CONCLUSIONS: PAR-4 expression is positively correlated with HCC; PAR-4 promotes malignant behavior of HCC cells mediated by the IL-6/STAT3 signaling pathway.

4.
Am J Hum Genet ; 107(2): 211-221, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32649856

RESUMO

Dual antiplatelet therapy reduces ischemic events in cardiovascular disease, but it increases bleeding risk. Thrombin receptors PAR1 and PAR4 are drug targets, but the role of thrombin in platelet aggregation remains largely unexplored in large populations. We performed a genome-wide association study (GWAS) of platelet aggregation in response to full-length thrombin, followed by clinical association analyses, Mendelian randomization, and functional characterization including iPSC-derived megakaryocyte and platelet experiments. We identified a single sentinel variant in the GRK5 locus (rs10886430-G, p = 3.0 × 10-42) associated with increased thrombin-induced platelet aggregation (ß = 0.70, SE = 0.05). We show that disruption of platelet GRK5 expression by rs10886430-G is associated with enhanced platelet reactivity. The proposed mechanism of a GATA1-driven megakaryocyte enhancer is confirmed in allele-specific experiments. Utilizing further data, we demonstrate that the allelic effect is highly platelet- and thrombin-specific and not likely due to effects on thrombin levels. The variant is associated with increased risk of cardiovascular disease outcomes in UK BioBank, most strongly with pulmonary embolism. The variant associates with increased risk of stroke in the MEGASTROKE, UK BioBank, and FinnGen studies. Mendelian randomization analyses in independent samples support a causal role for rs10886430-G in increasing risk for stroke, pulmonary embolism, and venous thromboembolism through its effect on thrombin-induced platelet reactivity. We demonstrate that G protein-coupled receptor kinase 5 (GRK5) promotes platelet activation specifically via PAR4 receptor signaling. GRK5 inhibitors in development for the treatment of heart failure and cancer could have platelet off-target deleterious effects. Common variants in GRK5 may modify clinical outcomes with PAR4 inhibitors, and upregulation of GRK5 activity or signaling in platelets may have therapeutic benefits.


Assuntos
Plaquetas/fisiologia , Doenças Cardiovasculares/genética , Receptores de Trombina/genética , Transdução de Sinais/genética , Trombina/genética , Alelos , Embolia/genética , Feminino , Estudo de Associação Genômica Ampla/métodos , Insuficiência Cardíaca/genética , Humanos , Pulmão/fisiologia , Masculino , Pessoa de Meia-Idade , Neoplasias/genética , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Receptor PAR-1/genética , Acidente Vascular Cerebral/genética
5.
Platelets ; 34(1): 2222846, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37394920

RESUMO

BMS-986141 is a novel, oral, protease-activated, receptor 4 (PAR4)-antagonist that exhibited robust antithrombotic activity and low bleeding risk in preclinical studies. The pharmacokinetic, pharmacodynamic, and tolerability profiles of BMS-986141 in healthy participants were assessed in a randomized, double-blind, placebo-controlled, single-ascending-dose (SAD; N = 60) study; a multiple-ascending-dose (MAD; N = 32) study; and a Japanese MAD (JMAD; N = 32) study. Exposure was dose-proportional for BMS-986141 2.5 mg and 150 mg; maximum concentrations were 17.6 ng/mL and 958 ng/mL; and areas under the curve (AUC) to infinity were 183 h* × ng/mL and 9207 h* × ng/mL, respectively. Mean half-life ranged from 33.7 to 44.7 hours across dose panels. The accumulation index following once-daily administration for 7 days suggested a 1.3- to 2-fold AUC increase at steady state. In the SAD study, BMS-986141 75 and 150 mg produced ≥80% inhibition of 25-100 µM PAR4 agonist peptide (AP)-induced platelet aggregation, without affecting PAR1-AP-induced platelet aggregation, through ≥24 hours postdose. In the MAD and JMAD studies, BMS-986141 doses ≥10 mg completely inhibited 12.5 µM and 25 µM PAR4-AP-induced platelet aggregation through 24 hours. This study found BMS-986141 was safe and well tolerated, with dose-proportional pharmacokinetics and concentration-dependent pharmacodynamics in healthy participants over a wide dose range. ClinicalTrials.gov ID: NCT02341638.


Why was the study done? Antiplatelet therapies have shortcomings that limit their clinical utility, and there is an unmet need for a new, safe, and effective antiplatelet agent with reduced bleeding risk.PAR4 antagonists are a promising novel class of antiplatelet drugs due to late-stage inhibition of thrombus growth with minimal effects on platelet-driven hemostasis.BMS-986141 is a novel, potent, orally bioavailable, small-molecule antagonist specific for PAR4.What is new? BMS-986141 is safe and well tolerated, with dose-proportional pharmacokinetics and concentration-dependent pharmacodynamics in healthy participants over a wide dose range.BMS-986141 has robust antithrombotic activity and low bleeding risk.What is the impact? BMS-986141 has the potential to improve the benefit­risk of antiplatelet therapy in patients with atherothrombosis.


Assuntos
Inibidores da Agregação Plaquetária , Agregação Plaquetária , Humanos , Administração Oral , Área Sob a Curva , Relação Dose-Resposta a Droga , Método Duplo-Cego , Voluntários Saudáveis , Hemorragia/induzido quimicamente
6.
Int J Mol Sci ; 24(9)2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37176005

RESUMO

Protease-activated receptors (PARs) are a class of integral membrane proteins that are cleaved by a variety of proteases, most notably thrombin, to reveal a tethered ligand and promote activation. PARs are critical mediators of platelet function in hemostasis and thrombosis, and therefore are attractive targets for anti-platelet therapies. Animal models studying platelet PAR physiology have relied heavily on genetically modified mouse strains, which have provided ample insight but have some inherent limitations. The current review aims to summarize the notable PAR expression and functional differences between the mouse and human, in addition to highlighting some recently developed tools to further study human physiology in mouse models.


Assuntos
Receptores Ativados por Proteinase , Receptores de Trombina , Humanos , Camundongos , Animais , Receptores Ativados por Proteinase/metabolismo , Receptores de Trombina/metabolismo , Especificidade da Espécie , Plaquetas/metabolismo , Trombina/metabolismo
7.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054966

RESUMO

Thrombin stimulates platelets via a dual receptor system of protease-activated receptors (PARs): PAR1 and PAR4. PAR1 activation induces a rapid and transient signal associated with the initiation of platelet aggregation, whereas PAR4 activation results in a prolonged signal, required for later phases, that regulates the stable formation of thrombus. In this study, we observed differential signaling pathways for thrombin-induced PAR1 and PAR4 activation in a human megakaryoblastic leukemia cell line, MEG-01. Interestingly, thrombin induced both calcium signaling and morphological changes in MEG-01 cells via the activation of PAR1 and PAR4, and these intracellular events were very similar to those observed in platelets shown in previous studies. We developed a novel image-based assay to quantitatively measure the morphological changes in living cells, and observed the underlying mechanism for PAR1- and PAR4-mediated morphological changes in MEG-01 cells. Selective inhibition of PAR1 and PAR4 by vorapaxar and BMS-986120, respectively, showed that thrombin-induced morphological changes were primarily mediated by PAR4 activation. Treatment of a set of kinase inhibitors and 2-aminoethoxydiphenyl borate (2-APB) revealed that thrombin-mediated morphological changes were primarily regulated by calcium-independent pathways and PAR4 activation-induced PI3K/Akt and RhoA/ROCK signaling pathways in MEG-01 cells. These results indicate the importance of PAR4-mediated signaling pathways in thrombin-induced morphological changes in MEG-01 cells and provide a useful in vitro cellular model for platelet research.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Trombina/metabolismo , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Plaquetas/metabolismo , Cálcio/metabolismo , Linhagem Celular , Imunofluorescência , Expressão Gênica , Humanos , Megacariócitos/citologia , Megacariócitos/metabolismo , Modelos Biológicos , Trombina/metabolismo
8.
J Biol Chem ; 295(8): 2520-2540, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-31892516

RESUMO

Proteinase-activated receptor (PAR)-4 is a member of the proteolytically-activated PAR family of G-protein-coupled receptors (GPCR) that represents an important target in the development of anti-platelet therapeutics. PARs are activated by proteolytic cleavage of their receptor N terminus by enzymes such as thrombin, trypsin, and cathepsin-G. This reveals the receptor-activating motif, termed the tethered ligand that binds intramolecularly to the receptor and triggers signaling. However, PARs are also activated by exogenous application of synthetic peptides derived from the tethered-ligand sequence. To better understand the molecular basis for PAR4-dependent signaling, we examined PAR4-signaling responses to a peptide library derived from the canonical PAR4-agonist peptide, AYPGKF-NH2, and we monitored activation of the Gαq/11-coupled calcium-signaling pathway, ß-arrestin recruitment, and mitogen-activated protein kinase (MAPK) pathway activation. We identified peptides that are poor activators of PAR4-dependent calcium signaling but were fully competent in recruiting ß-arrestin-1 and -2. Peptides that were unable to stimulate PAR4-dependent calcium signaling could not trigger MAPK activation. Using in silico docking and site-directed mutagenesis, we identified Asp230 in the extracellular loop-2 as being critical for PAR4 activation by both agonist peptide and the tethered ligand. Probing the consequence of biased signaling on platelet activation, we found that a peptide that cannot activate calcium signaling fails to cause platelet aggregation, whereas a peptide that is able to stimulate calcium signaling and is more potent for ß-arrestin recruitment triggered greater levels of platelet aggregation compared with the canonical PAR4 agonist peptide. These findings uncover molecular determinants critical for agonist binding and biased signaling through PAR4.


Assuntos
Receptores de Trombina/metabolismo , Transdução de Sinais , Trombina/metabolismo , Alanina/genética , Substituição de Aminoácidos , Cálcio/metabolismo , Sinalização do Cálcio , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Isomerismo , Sistema de Sinalização das MAP Quinases , Metilação , Simulação de Acoplamento Molecular , Proteínas Mutantes/metabolismo , Mutação/genética , Peptídeos/metabolismo , Fosforilação , Agregação Plaquetária , Receptores de Trombina/agonistas , Homologia Estrutural de Proteína , beta-Arrestinas/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 40(11): 2678-2685, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32907366

RESUMO

OBJECTIVE: PAR (protease-activated receptor)-4 antagonism has antiplatelet effects under conditions of high shear stress. We aimed to establish whether PAR4 antagonism had additive antithrombotic activity in the presence of factor Xa inhibition in an ex vivo model of acute arterial injury. Approach and Results: Fifteen healthy volunteers (29±6 years, 7 women) completed a phase zero double-blind randomized controlled crossover trial. Ex vivo platelet activation, platelet aggregation, and thrombus formation were measured following blood perfusion of low shear and high shear stress chambers. Upstream of the chambers, extracorporeal blood was admixed with (1) vehicle, (2) low-dose apixaban (20 ng/mL), (3) high-dose apixaban (80 ng/mL), (4) BMS-986141 (400 ng/mL), (5) BMS-968141 and low-dose apixaban, or (6) BMS-968141 and high-dose apixaban in 6 sequential studies performed in random order. Compared with vehicle, BMS-986141 demonstrated selective inhibition of PAR4-AP (agonist peptide)-stimulated platelet aggregation, platelet-monocyte aggregates, and P-selectin expression (P≤0.01 for all). Total thrombus area was reduced under both low shear and high shear stress conditions for all drug infusions (P<0.0001 for all versus vehicle). BMS-968141 reduced total (≤44.4%) and platelet-rich (≤39.3%) thrombus area, whereas apixaban reduced total (≤42.9%) and fibrin-rich (≤31.6%) thrombus area. Combination of BMS-986141 with apixaban caused a further modest reduction in total thrombus area (9.6%-12.4%), especially under conditions of high shear stress (P≤0.027). CONCLUSIONS: In the presence of factor Xa inhibition, PAR4 antagonism with BMS-986141 further reduces thrombus formation, especially under conditions of high shear stress. This suggests the potential for additive efficacy of combination PAR4 antagonism and factor Xa inhibition in the prevention of atherothrombotic events.


Assuntos
Plaquetas/efeitos dos fármacos , Inibidores do Fator Xa/administração & dosagem , Fibrinolíticos/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Pirazóis/administração & dosagem , Piridonas/administração & dosagem , Receptores de Trombina/antagonistas & inibidores , Trombose/prevenção & controle , Adulto , Plaquetas/metabolismo , Método Duplo-Cego , Quimioterapia Combinada , Inibidores do Fator Xa/farmacocinética , Feminino , Fibrinolíticos/farmacocinética , Humanos , Masculino , Pirazóis/farmacocinética , Piridonas/farmacocinética , Receptores de Trombina/sangue , Transdução de Sinais , Trombose/sangue , Adulto Jovem
10.
Exp Cell Res ; 395(2): 112205, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32738347

RESUMO

PURPOSE: Colon cancer is a common caner with high death rate in the world. The study aimed to detect the effect and mechanism of long non-coding RNA (LncRNA) MIR503HG on colon cancer. METHODS: The MIR503HG expression was measured in colon cancer tissues and cell lines by qRT-PCR. The proliferation, apoptosis, migration and invasion of colon cancer cells were measured by MTT, flow-cytometry, wound healing and transwell assay. The protein expression of E-cadherin, N-cadherin, and Vimentin was detected by Western blot. The target relationships among MIR503HG, miR-107 and Par4 were predicted by StarBase and TargetScan, and verified by luciferase reporter and RNA pull-down assay. The xenograft tumor model was constructed in mice to verify the inhibitory effect of MIR503HG in vivo. RESULTS: The expression of MIR503HG was decreased in colon cancer tissues and cell lines. MIR503HG overexpression inhibited cell proliferation, migration and invasion, promoted cell apoptosis, down-regulated N-cadherin and Vimentin, and up-regulated E-cadherin in colon cancer. MIR503HG negatively regulated its target miR-107. MiR-107 overexpression reversed the anti-tumor effects of MIR503HG overexpression on colon cancer cells. Par4 was a target of miR-107, which was positively regulated by MIR503HG. The promoting effects of MIR503HG silencing on colon cancer cells were eliminated by Par4 overexpression. CONCLUSION: MIR503HG regulated Par4 via sponging miR-107 in colon cancer, which promoting a new idea for the treatment of colon cancer.


Assuntos
Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Colo/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação para Baixo , Humanos , Receptores de Trombina/metabolismo , Vimentina/metabolismo
11.
J Stroke Cerebrovasc Dis ; 30(3): 105547, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33360254

RESUMO

OBJECTIVES: The inhibitory effects of P2Y12 receptor antagonist on PAR1- and PAR4-activating peptide (AP)-induced platelet aggregation have not been fully elucidated. The present study aimed to investigate the inhibitory effects of P2Y12 receptor antagonist on PAR1- and PAR4-AP-induced platelet aggregation using platelet-rich plasma (PRP) from individuals including patients with stroke or transient ischemic attack (TIA). MATERIALS AND METHODS: PRP was given to 10 healthy individuals pretreated in vitro with cangrelor, then stimulated with adenosine diphosphate (ADP), PAR4-AP, or PAR1-AP. Moreover, 20 patients were enrolled from 148 consecutive patients with acute ischemic stroke or TIA admitted to our institute between December 2017 and April 2019. PRP obtained from each patient before and >7 days after initiation of clopidogrel was similarly stimulated with these agonists. Platelet aggregation was measured using an automatic coagulation analyzer in all participants. RESULTS: In healthy individuals, ADP- and PAR4-AP-induced platelet aggregations were significantly inhibited depending on the cangrelor concentration in vitro, while PAR1-AP-induced platelet aggregation was slightly inhibited. In patients with stroke or TIA, clopidogrel inhibited ADP-induced platelet aggregation at all concentrations, and significantly inhibited PAR4-AP-induced platelet aggregation at 50 µmol/L of PAR4-AP (p<0.05), especially in 5 patients who showed high reactivity to PAR4-AP. PAR1-AP-induced platelet aggregation was also slightly inhibited. CONCLUSIONS: We showed significant inhibitory effects on PAR4-AP-induced platelet aggregation by clopidogrel in patients with stroke or TIA who had high reactivity to PAR4-AP.


Assuntos
Plaquetas/efeitos dos fármacos , Clopidogrel/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Oligopeptídeos/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Difosfato de Adenosina/farmacologia , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Plaquetas/metabolismo , Estudos de Casos e Controles , Clopidogrel/efeitos adversos , Feminino , Humanos , Ataque Isquêmico Transitório/sangue , Ataque Isquêmico Transitório/diagnóstico , AVC Isquêmico/sangue , AVC Isquêmico/diagnóstico , Masculino , Pessoa de Meia-Idade , Inibidores da Agregação Plaquetária/efeitos adversos , Valor Preditivo dos Testes , Estudos Prospectivos , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Resultado do Tratamento
12.
Pancreatology ; 20(8): 1698-1710, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33039292

RESUMO

BACKGROUND: We recently reported prostate apoptosis response 4 (Par-4), a potential tumor suppressor protein restrains epithelial-mesenchymal transition (EMT) properties and promotes mesenchymal-epithelial transition (MET) in invasive cancer cells by repressing Twist-1 promoter activity. Here, we demonstrate that genetic as well as pharmacological modulation of Par-4 by NGD16 (a small molecule antimetastatic agent), limits EMT-induced chemoresistance in aggressive cancer cells by suppressing MDM-2, a downstream effector of Twist-1. METHODS: Matrigel invasion assay, gelatin degradation assay, cell scattering assay, MTT assay and colony formation assay were used to study the proliferation and migration abilities of invasive cancer cells. Immunoblotting, immunocytochemistry, and immunoprecipitation analysis were utilized for determining protein expression and protein-protein interaction. 4T1 aggressive mouse carcinoma model was employed to evaluate tumor growth and lung metastasis. RESULTS: Treatment of gemcitabine (nucleoside analogue anticancer agent) to pancreatic cancer (Panc-1, MiaPaca-2) and breast cancer (MDA-MB-231) cells amplified MDM-2 expression along with increase in EMT properties. Conversely, NGD16 boosted expression of tumor suppressor Par-4 and inhibited invasion and migration abilities of these cells. Moreover, induction of Par-4 effectively diminished MDM-2 along with pro-EMT markers, whereas, augmented the expression of epithelial markers. Furthermore, siRNA-mediated silencing of Par-4 divulged that NGD16 exerts its EMT inhibitory effects in a Par-4-dependent manner. Mechanistically, Par-4 activation provokes p53 by disrupting MDM-2-p53 interaction, which restored epithelial characteristics in cancer cells. Additionally, partial knockdown of MDM-2 through siRNA pronounced the anti-proliferative and anti-invasive effects of NGD16. Finally, NGD16 efficiently inhibited tumor growth and lung metastasis in mouse mammary carcinoma model without showing any undesirable effects. CONCLUSION: Our findings unveil Par-4 as a key therapeutic target and NGD16 (the pharmacological modulator of Par-4) are potential tools to suppress EMT and associated chemoresistance, which could be exploited clinically for the treatment of aggressive cancers.


Assuntos
Neoplasias da Mama , Neoplasias Pancreáticas , Animais , Antineoplásicos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Movimento Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Receptores de Trombina , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
13.
Proc Natl Acad Sci U S A ; 114(41): 10948-10953, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28973891

RESUMO

C4a is a small protein released from complement component C4 upon activation of the complement system's classical and lectin pathways, which are important constituents of innate immune surveillance. Despite the structural similarity between C4a and well-described anaphylatoxins C3a and C5a, the binding partner and biological function of C4a have remained elusive. Using a cell-based reporter assay, we screened C4a against a panel of both known and orphan G protein-coupled receptors and now provide evidence that C4a is a ligand for protease-activated receptor (PAR)1 and PAR4. Whereas C4a showed no activity toward known anaphylatoxin receptors, it acted as an agonist for both PAR1 and PAR4 with nanomolar activity. In human endothelial cells, ERK activation by C4a was mediated through both PAR1 and PAR4 in a Gαi-independent signaling pathway. Like other PAR1 activators, C4a induced calcium mobilization through the PAR1/Gαq/PLCß signaling axis. Moreover, C4a increased stress fiber formation and enhanced endothelial permeability, both of which were reduced by PAR1 antagonists. In sum, our study identifies C4a as an untethered agonist for PAR1 and PAR4 with effects on cellular activation and endothelial permeability, thereby revealing another instance of cross-talk between the complement system and other host defense pathways.


Assuntos
Cálcio/metabolismo , Ativação do Complemento , Complemento C4a/metabolismo , Endotélio Vascular/metabolismo , Receptor PAR-1/agonistas , Receptores de Trombina/agonistas , Células Cultivadas , Complemento C4a/genética , Endotélio Vascular/citologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor PAR-1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Trombina/metabolismo , Transdução de Sinais
14.
Am J Physiol Cell Physiol ; 316(2): C264-C273, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30462538

RESUMO

Cancer-associated thrombosis is a common first presenting sign of malignancy and is currently the second leading cause of death in cancer patients after their malignancy. However, the molecular mechanisms underlying cancer-associated thrombosis remain undefined. In this study, we aimed to develop a better understanding of how cancer cells affect the coagulation cascade and platelet activation to induce a prothrombotic phenotype. Our results show that colon cancer cells trigger platelet activation in a manner dependent on cancer cell tissue factor (TF) expression, thrombin generation, activation of the protease-activated receptor 4 (PAR4) on platelets and consequent release of ADP and thromboxane A2. Platelet-colon cancer cell interactions potentiated the release of platelet-derived extracellular vesicles (EVs) rather than cancer cell-derived EVs. Our data show that single colon cancer cells were capable of recruiting and activating platelets and generating fibrin in plasma under shear flow. Finally, in a retrospective analysis of colon cancer patients, we found that the number of venous thromboembolism events was 4.5 times higher in colon cancer patients than in a control population. In conclusion, our data suggest that platelet-cancer cell interactions and perhaps platelet procoagulant EVs may contribute to the prothrombotic phenotype of colon cancer patients. Our work may provide rationale for targeting platelet-cancer cell interactions with PAR4 antagonists together with aspirin and/or ADP receptor antagonists as a potential intervention to limit cancer-associated thrombosis, balancing safety with efficacy.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/fisiologia , Neoplasias do Colo/sangue , Trombose/sangue , Plaquetas/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Estudos Transversais , Humanos , Estudos Retrospectivos , Trombose/patologia
15.
Breast Cancer Res Treat ; 177(2): 307-323, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31175498

RESUMO

PURPOSE: More than 90% of the breast cancer deaths occur due to the metastasis of the cancer cells to secondary organ sites. Increased Glucose-regulated protein 78 (GRP78) expression is critical for epithelial-mesenchymal transition (EMT) and invasion in breast cancer resulting in poor patient survival outcomes. Therefore, there is an urgent need of potential inhibitors of GRP78 for the abrogation of invasion and metastasis in breast cancer. METHODS: We investigated the effect of IKM5 (2-(1-(1H-indol-3-yl)octyl)-3-hydroxy-6-(hydroxymethyl)-4H-pyran-4-one) (a novel Indolylkojyl methane analogue) on invasion abilities of human breast cancer cells employing invadopodia formation, Matrigel invasion assays, and mouse models for metastasis. The mechanism underlying the anti-invasive effect of IKM5 was examined through molecular docking, immunoblotting, immunocytochemistry, co-immunoprecipitation analysis, siRNA silencing, and sub-cellular fractionation studies. RESULTS: Treatment with IKM5 at its sub-toxic concentration (200 nM) suppressed invasion and invadopodia formation, and growth factor-induced cell scattering of aggressive human breast cancer MDA-MB-231, MDA-MB-468, and MCF7 cells. IKM5 spontaneously binds to GRP78 (Ki = 1.35 µM) and downregulates its expression along with the EMT markers MMP-2, Twist1, and Vimentin. Furthermore, IKM5 amplified the expression and nuclear translocation of tumor suppressor Par-4 to control NF-kB-mediated pro-EMT activities. Interestingly, IKM5 disrupts the interaction between GRP78 and TIMP-1 by inhibiting GRP78 in a Par-4-dependent manner. Moreover, IKM5 inhibited tumor growth and lung metastasis at a safe dose of 30 mg/kg/body weight. CONCLUSION: Our study warrants IKM5, a potential anticancer agent that can abrogate invasion and metastasis, suggesting its clinical development for the treatment of patients with advanced breast cancer.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Choque Térmico/genética , Metano/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Chaperona BiP do Retículo Endoplasmático , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Metaloproteinases da Matriz , Metano/análogos & derivados , Metano/química , Metano/farmacocinética , Camundongos , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Transporte Proteico , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Anat ; 234(6): 830-838, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30861119

RESUMO

Human salivary gland (SG) branching morphogenesis is an intricate mechanism divided into stages, prebud, initial bud, pseudoglandular, canalicular, and terminal bud, to form the final lobular structure of the organ. The coordination of molecular cascades, including cell proliferation and apoptosis, are fundamental to this process. The intrinsic apoptosis pathway appears to be important in the early phases of ductal cavitation and luminisation; however, the role of the extrinsic apoptosis pathway has still to be determined. Questions remain as to whether the latter mechanism participates in the maintenance of the ductal lumen; therefore, the present study investigated the expression of proteins Prostate apoptosis response-4 (Par-4), Fas cell surface death receptor (Fas), Fas ligand (FasL), pleckstrin homology-like domain family A member 1 (PHLDA1), caspase-3, B-cell CLL/lymphoma 2 (Bcl-2), survivin, Ki-67, mucin 1 (MUC1), and secreted protein acidic and cysteine-rich (SPARC) during distinct phases of human SG development (50 specimens). This strategy aimed to draw an immunomorphological map of the proteins involved in apoptosis, cell proliferation, and tissue maturation during the SG branching morphogenesis process. Par-4 was positive at all stages except the pre-acinar phase. Fas and FasL were expressed in few cells. PHLDA1 was expressed in all phases but not in the terminal bud. Bcl-2 expression was mainly negative (expressed in few cells). Survivin showed a cytoplasmic expression pattern in the early phases of development, which changed to a predominantly nuclear expression during development into more differentiated structures. Ki-67 was expressed mainly at the pseudoglandular stage. MUC1 was positive in the pseudoglandular stage with a cytoplasmic pattern in regions of early luminal opening. Immunostaining for SPARC and caspase-3 was negative. Our results suggest that proteins associated with the regulation of extrinsic and intrinsic apoptosis contribute to apoptosis during specific phases of the early formation of SGs in humans.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Glândulas Salivares/embriologia , Embrião de Mamíferos , Feto , Humanos , Organogênese/fisiologia
17.
Toxicol Appl Pharmacol ; 385: 114773, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31678245

RESUMO

Tumor associated macrophages (TAMs) have a crucial role in cancer progression, metastasis and drug response. Piroxicam and sulindac sulfide are non-steroidal anti-inflammatory drugs (NSAID) that decrease the incidence and progression of several types of cancer. However, their role in suppressing the interactions between TAMs and cancer cells remain unclear. Herein, we studied the impact of human monocytes conditioned media (CM) on cellular proliferation of ER-dependent MCF-7 and ER-independent MDA-MB-231 cells, and the effects of piroxicam and sulindac sulfide on the expression levels of RAS, COX-2, IL-6, IL-1ß and PAR-4 (qRT-PCR), BCL-2 and BAX (western blot), Caspase-3, VEGF-a and PGE2 (ELISA), MMP-2 and -9 (zymography) in the stimulated cells. Our results showed that CM caused a significant increase in cells survival through significant increase in RAS expression which resulted in upregulation of COX-2, PGE2, BCL-2, IL-6, IL-1ß, VEGF-A and MMP-9 and down regulation of PAR-4. Treatment with one of the NSAIDs used in this study produced a time and concentration dependent growth inhibition of stimulated cells by inhibiting RAS expression. Suppression of RAS was accompanied by downregulation of its downstream signaling of IL-1ß, IL-6, COX-2 and PGE2, activation of apoptotic machinery through upregulation of PAR-4 and caspase-3, as well as, inhibition of BCL-2, VEGF-A, MMP-2 and MMP-9. In conclusion, our data support the role of piroxicam and sulindac sulfide in suppressing inflammation-driven breast cancer progression and identifies promising novel target in RAS and PAR-4 signaling.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Neoplasias da Mama/patologia , Inflamação/prevenção & controle , Macrófagos/fisiologia , Proteínas ras/fisiologia , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dinoprostona/biossíntese , Feminino , Humanos , Interleucina-1beta/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Transdução de Sinais/fisiologia , Proteínas ras/antagonistas & inibidores
18.
Bioorg Med Chem ; 27(1): 116-124, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30503410

RESUMO

BMS-986120 is a PAR4 antagonist that is being investigated as an antiplatelet agent in phase I clinical trial. An improved synthesis of BMS-986120 has been developed. Based on the novel synthetic approach to BMS-986120, a series of deuterated derivatives of BMS-986120 have been synthesized and biologically evaluated to search for more potent antiplatelet agents. The in vitro antiplatelet assay by turbidimetry demonstrated that PC-2 and PC-6 had IC50 values of 6.30 nM and 6.97 nM, respectively, versus BMS-986120 with an IC50 of 7.80 nM. The result of in vitro metabolic stability study showed that all of the deuterated compounds had similar half-life (T1/2) and intrinsic clearance (Clint) in comparison with BMS-986120. Further probing the metabolic profile of BMS-986120 is worth being conducted.


Assuntos
Benzofuranos/farmacologia , Imidazóis/farmacologia , Morfolinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Trombina/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Benzofuranos/síntese química , Benzofuranos/química , Plaquetas/efeitos dos fármacos , Deutério , Estabilidade de Medicamentos , Humanos , Imidazóis/síntese química , Imidazóis/química , Masculino , Camundongos Endogâmicos ICR , Microssomos Hepáticos/metabolismo , Morfolinas/síntese química , Morfolinas/química , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/síntese química , Inibidores da Agregação Plaquetária/química , Tiazóis/síntese química , Tiazóis/química
19.
Platelets ; 30(1): 126-135, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30560697

RESUMO

Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.


Assuntos
Plaquetas/metabolismo , Comunicação Celular , Grânulos Citoplasmáticos/metabolismo , Leucócitos/metabolismo , Receptores de Trombina/metabolismo , Animais , Biomarcadores , Citometria de Fluxo , Humanos , Masculino , Papio , Ativação Plaquetária , Agregação Plaquetária
20.
BMC Genomics ; 19(1): 462, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29907081

RESUMO

BACKGROUND: Caenorhabditis elegans can endure long periods of environmental stress by altering their development to execute a quiescent state called "dauer". Previous work has implicated LKB1 - the causative gene in the autosomal dominant, cancer pre-disposing disease called Peutz-Jeghers Syndrome (PJS), and its downstream target AMPK, in the establishment of germline stem cell (GSC) quiescence during the dauer stage. Loss of function mutations in both LKB1/par-4 and AMPK/aak(0) result in untimely GSC proliferation during the onset of the dauer stage, although the molecular mechanism through which these factors regulate quiescence remains unclear. Curiously, the hyperplasia observed in par-4 mutants is more severe than AMPK-compromised dauer larvae, suggesting that par-4 has alternative downstream targets in addition to AMPK to regulate germline quiescence. RESULTS: We conducted three genome-wide RNAi screens to identify potential downstream targets of the protein kinases PAR-4 and AMPK that mediate dauer-dependent GSC quiescence. First, we screened to identify genes that phenocopy the par-4-dependent hyperplasia when compromised by RNAi. Two additional RNAi screens were performed to identify genes that suppressed the germline hyperplasia in par-4 and aak(0) dauer larvae, respectively. Interestingly, a subset of the candidates we identified are involved in the regulation of cell polarity and cytoskeletal function downstream of par-4, in an AMPK-independent manner. Moreover, we show that par-4 temporally regulates actin cytoskeletal organization within the dauer germ line at the rachis-adjacent membrane, in an AMPK-independent manner. CONCLUSION: Our data suggest that the regulation of the cytoskeleton and cell polarity may contribute significantly to the tumour suppressor function of LKB1/par-4.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Proteínas de Caenorhabditis elegans/genética , Células Germinativas/citologia , Proteínas Serina-Treonina Quinases/genética , Células-Tronco/citologia , Quinases Proteína-Quinases Ativadas por AMP , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/ultraestrutura , Polaridade Celular/genética , Citoesqueleto , Genoma , Células Germinativas/ultraestrutura , Hiperplasia , Larva/citologia , Larva/genética , Larva/ultraestrutura , Mutação , Proteínas Quinases/genética , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa