Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 529
Filtrar
1.
Int J Hematol ; 119(2): 196-204, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38228939

RESUMEN

The protein C (PC) pathway involves physiological anticoagulant factors (PC, protein S [PS], and factor V) and performs major anticoagulant functions in adults. Variations in overall PC pathway function due to dynamic changes in PC and PS in early childhood are poorly understood. We aimed to evaluate the contributions of PC pathway function during early childhood by measuring changes in plasma thrombin generation (TG) after administration of the PC activator protac. We evaluated correlations between anticoagulant factors and percentage of protac-induced coagulation inhibition (PiCi%). Before protac addition, TG in newborns (n = 35), infants (n = 42), young children (n = 35), and adults (n = 20) were 525 ± 74, 720 ± 96, 785 ± 53, and 802 ± 64 mOD/min, and PiCi% were 42.1 ± 9.9, 69.8 ± 11.0, 82.9 ± 4.4, and 86.9 ± 3.4%, respectively. The distribution of PiCi% on the two axes of TG (with or without protac) changed continuously with age and differed from that of warfarin-treated plasma and adult PC- or PS-deficient plasma. PiCi% increased dynamically during infancy and correlated with PS levels in newborns and PC levels in young children. Addition of PC or fresh frozen plasma equivalent to approximately 25% PC to PC-deficient plasma improved PiCi%. This automatic measurement requires only a small sample volume and is useful for analysis of developmental hemostasis.


Asunto(s)
Proteína C , Quimera Dirigida a la Proteólisis , Adulto , Niño , Preescolar , Humanos , Recién Nacido , Anticoagulantes/farmacología , Antitrombinas/farmacología , Coagulación Sanguínea , Proteína C/análisis , Proteína C/metabolismo , Proteína C/farmacología , Proteína S/metabolismo , Trombina/metabolismo , Lactante
2.
J Trauma Acute Care Surg ; 96(1): 116-122, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37733304

RESUMEN

BACKGROUND: Activated Protein C (aPC) plays dual roles after injury, driving both trauma-induced coagulopathy (TIC) by cleaving, and thus inactivating, factors Va and VIIIa and depressing fibrinolysis while also mediating an inflammomodulatory milieu via protease activated receptor-1 (PAR-1) cytoprotective signaling. Because of this dual role, it represents and ideal target for study and therapeutics after trauma. A known aPC variant, 3K3A-aPC, has been engineered to preserve cytoprotective activity while retaining minimal anticoagulant activity rendering it potentially ideal as a cytoprotective therapeutic after trauma. We hypothesized that 3K3A-aPC would mitigate the endotheliopathy of trauma by protecting against endothelial permeability. METHODS: We used electric cell-substrate impedance sensing to measure permeability changes in real time in primary endothelial cells. These were cultured, grown to confluence, and treated with a 2 µg/mL solution of 3K3A-aPC at 180 minutes, 120 minutes, 60 minutes, 30 minutes prior to stimulation with ex vivo plasma taken from severely injured trauma patients (Injury Severity Score > 15 and BD < -6) (trauma plasma [TP]). Cells treated with thrombin and untreated cells were included in this study as control groups. Permeability changes were recorded in real time via electric cell-substrate impedance sensing for 30 minutes after treatment with TP. We quantified permeability changes in the control and treatment groups as area under the curve (AUC). Rac1/RhoA activity was also compared between these groups. Statistical significance was determined by one-way ANOVA followed by a post hoc analysis using Tukey's multiple comparison's test. RESULTS: Treatment with aPC mitigated endothelial permeability induced by ex vivo trauma plasma at all pre-treatment time points. The AUC of the 30-minute 3K3A-aPC pretreatment group was higher than TP alone (mean diff. 22.12 95% CI [13.75, 30.49], p < 0.0001) (Figure). Moreover, the AUC of the 60-minute, 120-minute, and 180-minute pretreatment groups was also higher than TP alone (mean diff., 16.30; 95% confidence interval [CI], 7.93-24.67; 19.43; 95% CI, 11.06-27.80, and 18.65; 95% CI, 10.28-27.02;, all p < 0.0001, respectively). Rac1/RhoA activity was higher in the aPC pretreatment group when compared with all other groups ( p < 0.01). CONCLUSION: Pretreatment with 3K3A-aPC, which retains its cytoprotective function but has only ~5% of its anticoagulant function, abrogates the effects of trauma-induced endotheliopathy. This represents a potential therapeutic treatment for dysregulated thromboinflammation for injured patients by minimizing aPC's role in trauma-induced coagulopathy while concurrently amplifying its essential cytoprotective function. LEVEL OF EVIDENCE: Prognostic and Epidemiological; Level III.


Asunto(s)
Proteína C , Trombosis , Humanos , Proteína C/farmacología , Proteína C/uso terapéutico , Proteína C/metabolismo , Células Endoteliales/metabolismo , Tromboinflamación , Inflamación/metabolismo , Anticoagulantes/uso terapéutico
3.
J Trauma Acute Care Surg ; 96(2): 276-286, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37335129

RESUMEN

BACKGROUND: Activated protein C (APC) is one of the mechanisms contributing to coagulopathy, which is associated with high mortality. The counteraction of the APC pathway could help ameliorate bleeding. However, patients also transform frequently from a hemorrhagic state to a prothrombotic state at a later time. Therefore, a prohemostatic therapeutic intervention should take this thrombotic risk into consideration. OBJECTIVES: CT-001 is a novel factor VIIa (FVIIa) with enhanced activity and desialylated N-glycans for rapid clearance. We assessed CT-001 clearance in multiple species and its ability to reverse APC-mediated coagulopathic blood loss. METHODS: The N-glycans on CT-001 were characterized by liquid chromatography-mass spectrometry. Three species were used to evaluate the pharmacokinetics of the molecule. The potency and efficacy of CT-001 under APC pathway-induced coagulopathic conditions were assessed by coagulation assays and bleeding models. RESULTS: The N-glycosylation sites of CT-001 had high occupancy of desialylated N-glycans. CT-001 exhibited 5 to 16 times higher plasma clearance in human tissue factor knockin mice, rats, and cynomolgus monkeys than wildtype FVIIa. CT-001 corrected the activated partial thromboplastin time and thrombin generation of coagulopathic plasma to normal in in vitro studies. In an APC-mediated saphenous vein bleeding model, 3 mg/kg of CT-001 reduced bleeding time in comparison with wildtype FVIIa. The correction of bleeding by CT-001 was also observed in a coagulopathic tail amputation severe hemorrhage mouse model. The efficacy of CT-001 is independent of the presence of tranexamic acid, and the combination of CT-001 and tranexamic acid does not lead to increased thrombogenicity. CONCLUSION: CT-001 corrected APC pathway-mediated coagulopathic conditions in preclinical studies and could be a potentially safe and effective procoagulant agent for addressing APC-mediated bleeding.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Ácido Tranexámico , Humanos , Ratones , Ratas , Animales , Proteína C/farmacología , Proteína C/uso terapéutico , Ácido Tranexámico/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea/etiología , Hemostasis , Hemorragia , Factor VIIa/uso terapéutico , Factor VIIa/farmacología , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Tomografía Computarizada por Rayos X
4.
Wound Repair Regen ; 32(1): 90-103, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38155595

RESUMEN

Various preclinical and clinical studies have demonstrated the robust wound healing capacity of the natural anticoagulant activated protein C (APC). A bioengineered APC variant designated 3K3A-APC retains APC's cytoprotective cell signalling actions with <10% anticoagulant activity. This study was aimed to provide preclinical evidence that 3K3A-APC is efficacious and safe as a wound healing agent. 3K3A-APC, like wild-type APC, demonstrated positive effects on proliferation of human skin cells (keratinocytes, endothelial cells and fibroblasts). Similarly it also increased matrix metollaproteinase-2 activation in keratinocytes and fibroblasts. Topical 3K3A-APC treatment at 10 or 30 µg both accelerated mouse wound healing when culled on Day 11. And at 10 µg, it was superior to APC and had half the dermal wound gape compared to control. Further testing was conducted in excisional porcine wounds due to their congruence to human skin. Here, 3K3A-APC advanced macroscopic healing in a dose-dependent manner (100, 250 and 500 µg) when culled on Day 21. This was histologically corroborated by greater collagen maturity, suggesting more advanced remodelling. A non-interference arm of this study found no evidence that topical 3K3A-APC caused either any significant systemic side-effects or any significant leakage into the circulation. However the female pigs exhibited transient and mild local reactions after treatments in week three, which did not impact healing. Overall these preclinical studies support the hypothesis that 3K3A-APC merits future human wound studies.


Asunto(s)
Células Endoteliales , Proteína C , Femenino , Humanos , Animales , Ratones , Porcinos , Proteína C/farmacología , Proteína C/metabolismo , Proteína C/uso terapéutico , Células Endoteliales/metabolismo , Cicatrización de Heridas , Fibrinolíticos/uso terapéutico , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico
5.
Int J Mol Sci ; 24(13)2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37445820

RESUMEN

3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with cytoprotective properties and reduced bleeding risks. We studied the potential use of 3K3A-APC as a multi-target therapeutic option for choroidal neovascularization (CNV), a common cause of vision loss in age-related macular degeneration. CNV was induced by laser photocoagulation in a murine model, and 3K3A-APC was intravitreally injected. The impact of 3K3A-APC treatment on myeloid and microglia cell activation and recruitment and on NLRP3 inflammasome, IL-1ß, and VEGF levels was assessed using cryosection, retinal flat-mount immunohistochemistry and vascular imaging. Additionally, we evaluated the use of fluorescein angiography as a surrogate marker for in vivo evaluation of the efficacy of 3K3A-APC treatment against leaking CNV lesions. Our results demonstrated that 3K3A-APC treatment significantly reduced the accumulation and activation of myeloid cells and microglia in the CNV area and decreased the NLRP3 and IL-1ß levels at the CNV site and the surrounding retina. Furthermore, 3K3A-APC treatment resulted in leakage regression and CNV growth suppression. These findings indicate that the anti-inflammatory activities of 3K3A-APC contribute to CNV inhibition. Our study suggests the potential use of 3K3A-APC as a novel multi-target treatment for CNV.


Asunto(s)
Neovascularización Coroidal , Proteína C , Ratones , Animales , Proteína C/farmacología , Proteína C/uso terapéutico , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Factor A de Crecimiento Endotelial Vascular , Retina/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
6.
J Matern Fetal Neonatal Med ; 36(1): 2183467, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36935364

RESUMEN

BACKGROUND: Inflammation is associated with many disorders of preterm infants including periventricular leukomalacia, chronic lung disease, and necrotizing enterocolitis. Activated protein c (APC) has shown positive immunomodulatory effects. OBJECTIVES: We aimed to study neutrophil and monocyte function in response to lipopolysaccharide (LPS) and APC stimulation ex vivo in preterm infants <32 weeks gestation over the first week of life compared to neonatal and adult controls. METHODS: Peripheral blood was taken on day 1, 3, and 7 and stimulated with LPS in the absence or presence of APC. Expression of toll-like receptor 4 (TLR4) and CD11b and reactive oxygen intermediate (ROI) release from neutrophils and monocytes was examined by flow cytometry. RESULTS: LPS induced neutrophil ROI in adults and preterm infants and was significantly reduced by APC. Baseline and LPS-induced monocyte ROI production in preterm neonates was increased compared to adult and term controls. Neutrophil TLR4 baseline expression was higher in term controls compared to preterm infants. CONCLUSION: Increased systemic ROI release in preterm infants may mediate tissue damage, ROI was reduced by APC. However, due to the high risk of hemorrhage further examination of APC mutant forms with anti-inflammatory but decreased anticoagulant properties is merited.


Asunto(s)
Recien Nacido Prematuro , Neutrófilos , Adulto , Lactante , Recién Nacido , Humanos , Neutrófilos/metabolismo , Monocitos/metabolismo , Proteína C/metabolismo , Proteína C/farmacología , Receptor Toll-Like 4/metabolismo , Lipopolisacáridos/farmacología
7.
Int J Radiat Biol ; 99(7): 1109-1118, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36827630

RESUMEN

PURPOSE: The goal of the current study was to identify longitudinal changes in urinary metabolites following IR exposure and to determine potential alleviation of radiation toxicities by administration of recombinant APC formulations. MATERIALS AND METHODS: Female adult WAG/RijCmcr rats were irradiated with 13.0 Gy leg-out partial body X-rays; longitudinally collected urine samples were subject to LC-MS based metabolomic profiling. Sub-cohorts of rats were treated with three variants of recombinant APC namely, rat wildtype (WT) APC, rat 3K3A mutant form of APC, and human WT APC as two bolus injections at 24 and 48 hours post IR. RESULTS: Radiation induced robust changes in the urinary profiles leading to oxidative stress, severe dyslipidemia, and altered biosynthesis of PUFAs, glycerophospholipids, sphingolipids, and steroids. Alterations were observed in multiple metabolic pathways related to energy metabolism, nucleotide biosynthesis and metabolism that were indicative of disrupted mitochondrial function and DNA damage. On the other hand, sub-cohorts of rats that were treated with rat wildtype-APC showed alleviation of radiation toxicities, in part, at the 90-day time point, while rat 3K3A-APC showed partial alleviation of radiation induced metabolic alterations 14 days after irradiation. CONCLUSIONS: Taken together, these results show that augmenting the Protein C pathway and activity via administration of recombinant APC may be an effective approach for mitigation of radiation induced normal tissue toxicity.


Asunto(s)
Proteína C , Traumatismos por Radiación , Ratas , Animales , Femenino , Humanos , Proteína C/farmacología , Metaboloma , Metabolómica
8.
Tohoku J Exp Med ; 259(3): 209-219, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36543245

RESUMEN

The Holliday Junction-Recognition Protein (HJURP) was upregulated in several tumors, which was associated with poor outcome. This study investigated the effects of the HJURP-mediated c-Jun N-terminal kinase (JNK)/ signal transducer and activator of transcription 3 (STAT3) pathway on bladder urothelial carcinoma (BLUC). Online databases were used to analyze HJURP expression in BLUC and the correlation of HJURP to JNK1 [mitogen-activated protein kinase 8 (MAPK8)], JNK2 (MAPK9), STAT3, marker of proliferation Ki-67 (MKI67), proliferating cell nuclear antigen (PCNA), cyclin dependent kinase 2 (CDK2), CDK4 and CDK6. HJURP expression was detected in BLUC cells and human normal primary bladder epithelial cells (BdECs). BLUC cells were treated with HJURP lentivirus activation /shRNA lentivirus particles or JNK inhibitor SP600125. HJURP was upregulated in BLUC tissues and correlated with poor prognosis of patients (all P < 0.05). HJURP in tumor positively correlated with MAPK8 (R = 0.30), MAPK9 (R = 0.30), STAT3 (R = 0.15), MKI67 (R = 0.60), PCNA (R = 0.46), CDK2 (R = 0.39), CDK4 (R = 0.24) and CDK6 (R = 0.21). The JNK inhibitor SP600125 decreased p-JNK/JNK and p-STAT3/STAT3 in BLUC cells, which was reversed by HJURP overexpression (P < 0.05). The HJURP-mediated JNK/STAT3 pathway promoted BLUC cell proliferation and inhibited cell apoptosis (P < 0.05). HJURP reversed the arrested G0/G1 phase of BLUC cells by SP600125. HJURP acted as an oncogene to regulate BLUC cell proliferation, apoptosis and the cell cycle by mediating the JNK/STAT3 pathway. Therefore, HJURP targeting might be an attractive novel therapeutic target for early diagnosis and treatment in BLUC.


Asunto(s)
Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/farmacología , ADN Cruciforme , Proteína C/metabolismo , Proteína C/farmacología , Vejiga Urinaria , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Ciclo Celular , Proliferación Celular , Apoptosis
9.
Int J Mol Sci ; 23(22)2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36430674

RESUMEN

3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with pleiotropic cytoprotective properties albeit without the bleeding risks. The anti-inflammatory activities of 3K3A-APC were demonstrated in multiple preclinical injury models, including various neurological disorders. We determined the ability of 3K3A-APC to inhibit ocular inflammation in a murine model of lipopolysaccharide (LPS)-induced uveitis. Leukocyte recruitment, microglia activation, NLRP3 inflammasome and IL-1ß levels were assessed using flow cytometry, retinal cryosection histology, retinal flatmount immunohistochemistry and vascular imaging, with and without 3K3A-APC treatment. LPS triggered robust inflammatory cell recruitment in the posterior chamber. The 3K3A-APC treatment significantly decreased leukocyte numbers and inhibited leukocyte extravasation from blood vessels into the retinal parenchyma to a level similar to controls. Resident microglia, which underwent an inflammatory transition following LPS injection, remained quiescent in eyes treated with 3K3A-APC. An inflammation-associated increase in retinal thickness, observed in LPS-injected eyes, was diminished by 3K3A-APC treatment, suggesting its clinical relevancy. Finally, 3K3A-APC treatment inhibited inflammasome activation, determined by lower levels of NLRP3 and its downstream effector IL-1ß. Our results highlight the anti-inflammatory properties of 3K3A-APC in ocular inflammation and suggest its potential use as a novel treatment for retinal diseases associated with inflammation.


Asunto(s)
Oftalmopatías , Inflamasomas , Proteína C , Animales , Ratones , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Lipopolisacáridos/toxicidad , Microglía/efectos de los fármacos , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína C/farmacología , Proteína C/uso terapéutico , Oftalmopatías/tratamiento farmacológico , Oftalmopatías/patología
10.
Chem Biodivers ; 19(10): e202200494, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36198620

RESUMEN

Biofilm-producing Staphylococcus aureus (SA) strains are frequently found in medical environments, from surgical/ wound sites, medical devices. These biofilms reduce the efficacy of applied antibiotics during the treatment of several infections, such as cystic fibrosis, endocarditis, or urinary tract infections. Thus, the development of potential therapeutic agents to destroy the extra protective biofilm layers or to inhibit the biofilm-producing enzymes is urgently needed. Advanced and cost-effective bioinformatics tools are advantageous in locating and speeding up the selection of antibiofilm candidates. Based on the potential drug characteristics, we have selected one-hundred thirty-three antibacterial peptides derived from insects to assess for their antibiofilm potency via molecular docking against five putative biofilm formation and regulated target enzymes: the staphylococcal accessory regulator A or SarA (PDB ID: 2FRH), 4,4'-diapophytoene synthase or CrtM (PDB ID: 2ZCQ), clumping factor A or ClfA (PDB ID: 1N67) and serine-aspartate repeat protein C or SdrC (PDB ID: 6LXH) and sortase A or SrtA (PDB ID: 1T2W) of SA bacterium. In this study, molecular docking was performed using HPEPDOCK and HDOCK servers, and molecular interactions were examined using BIOVIA Discovery Studio Visualizer-2019. The docking score (kcal/mol) range of five promising antibiofilm peptides against five targets was recorded as follows: diptericin A (-215.52 to -303.31), defensin (-201.11 to -301.92), imcroporin (-212.08 to -287.64), mucroporin (-228.72 to -286.76), apidaecin II (-203.90 to -280.20). Among these five, imcroporin and mucroporin were 13 % each, while defensin contained only 1 % of positive net charged residues (Arg+Lys) projected through ProtParam and NetWheels tools. Similarly, imcroporin, mucroporin and apidaecin II were 50 %, while defensin carried 21.05 % of hydrophobic residues predicted by the tool PEPTIDE. 2.0. Most of the peptides exhibited potential characteristics to inhibit S. aureus-biofilm formation via disrupting the cell membrane and cytoplasmic integrity. In summary, the proposed hypothesis can be considered a cost-effective platform for selecting the most promising bioactive drug candidates within a limited timeframe with a greater chance of success in experimental and clinical studies.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Simulación del Acoplamiento Molecular , Proteína C/farmacología , Proteína C/uso terapéutico , Ácido Aspártico/farmacología , Ácido Aspártico/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Biopelículas , Antibacterianos/farmacología , Defensinas/farmacología , Defensinas/uso terapéutico , Insectos , Serina/farmacología , Serina/uso terapéutico , Pruebas de Sensibilidad Microbiana
11.
Infect Immun ; 90(10): e0037622, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36121221

RESUMEN

Acinetobacter baumannii is a formidable opportunistic pathogen that is notoriously difficult to eradicate from hospital settings. This resilience is often attributed to a proclivity for biofilm formation, which facilitates a higher tolerance toward external stress, desiccation, and antimicrobials. Despite this, little is known regarding the mechanisms orchestrating A. baumannii biofilm formation. Here, we performed RNA sequencing (RNA-seq) on biofilm and planktonic populations for the multidrug-resistant isolate AB5075 and identified 438 genes with altered expression. To assess the potential role of genes upregulated within biofilms, we tested the biofilm-forming capacity of their respective mutants from an A. baumannii transposon library. In so doing, we uncovered 24 genes whose disruption led to reduced biofilm formation. One such element, cold shock protein C (cspC), had a highly mucoid colony phenotype, enhanced tolerance to polysaccharide degradation, altered antibiotic tolerance, and diminished adherence to abiotic surfaces. RNA-seq of the cspC mutant revealed 201 genes with altered expression, including the downregulation of pili and fimbria genes and the upregulation of multidrug efflux pumps. Using transcriptional arrest assays, it appears that CspC mediates its effects, at least in part, through RNA chaperone activity, influencing the half-life of several important transcripts. Finally, we show that CspC is required for survival during challenge by the human immune system and is key for A. baumannii dissemination and/or colonization during systemic infection. Collectively, our work identifies a cadre of new biofilm-associated genes within A. baumannii and provides unique insight into the global regulatory network of this emerging human pathogen.


Asunto(s)
Acinetobacter baumannii , Humanos , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Biopelículas , Proteínas y Péptidos de Choque por Frío/genética , Proteínas y Péptidos de Choque por Frío/metabolismo , Polisacáridos/metabolismo , Proteína C/metabolismo , Proteína C/farmacología , ARN/metabolismo , Virulencia/genética
12.
J Ovarian Res ; 15(1): 69, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35668504

RESUMEN

BACKGROUND: Dysregulation of Ectonucleoside Triphospahate Diphosphohydrolase 5 (ENTPD5) in tumors might be associated with tumor progression, while the role of ENTPD5 in the growth and metastasis of serous ovarian cancer (SOC) is still unclear. METHODS: ENTPD5 expression patterns in ovarian cancer tissues were analyzed by qRT-PCR and immunohistochemistry assay (IHC). Two SOC cell lines, SKOV3 and OVCAR8, were stably transfected with lentivirus to build knockdown and overexpression cell lines. Clone formation assay, collagen gel droplet culture technology, wound healing assay and flow cytometry were used to assess the migration and growth traits of SOC cells. Expression levels of ENTPD5, glucose regulated protein 78 (GRP78), eukaryotic translation initiation factor 2 alpha (eIF-2α), phosphorylated -eIF-2α and, C/EBP homologous protein (CHOP) in SOC cells were detected by Western blot. RESULTS: Compared to fallopian tube tissues, the expression of ENTPD5 was significantly higher in tumor tissues obtained from SOC patients, and positively correlated with clinical stage and metastasis. ENTPD5 knockdown robustly inhibited cell proliferation, migration, whereas ENTPD5 overexpression elicited the opposite effect on SOC cells. ENTPD5 knockdown arrested cell cycle in G0/G1 phase and increased apoptosis. Importantly, ENTPD5 knockdown was associated with significantly decreased protein levels for GRP78, CHOP, and p-eIF-2α, suggesting possible involvement of ENTPD5 in endoplasmic reticulum stress (ERS). CONCLUSIONS: Our study demonstrates that ENTPD5 knockdown inhibited SOC cell proliferation, migration and restrained the activation of the GRP78/p-eIF-2α/CHOP pathway, which provides a potentially effective therapeutic target for the treatment of SOC.


Asunto(s)
Cistadenocarcinoma Seroso , Neoplasias Ováricas , Apoptosis , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Cistadenocarcinoma Seroso/patología , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/farmacología , Femenino , Glucosa , Humanos , Proteínas Oncogénicas , Neoplasias Ováricas/patología , Proteína C/farmacología , Pirofosfatasas/farmacología
13.
Blood Adv ; 6(11): 3304-3314, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35390147

RESUMEN

Hemophilia A and B are hereditary coagulation defects resulting in unstable blood clotting and recurrent bleeding. Current factor replacement therapies have major limitations such as the short half-life of the factors and development of inhibitors. Alternative approaches to rebalance the hemostasis by inhibiting the anticoagulant pathways have recently gained considerable interest. In this study, we tested the therapeutic potential of a monoclonal antibody, HAPC1573, that selectively blocks the anticoagulant activity of human activated protein C (APC). We generated F8-/- or F9-/- hemophilia mice expressing human protein C by genetically replacing the murine Proc gene with the human PROC. The resulting PROC+/+;F8-/- or PROC+/+;F9-/- mice had bleeding characteristics similar to their corresponding F8-/- or F9-/- mice. Pretreating the PROC+/+;F8-/- mice with HAPC1573 shortened the tail bleeding time. HAPC1573 pretreatment significantly reduced mortality and alleviated joint swelling, similar to those treated with either FVIII or FIX, of either PROC+/+;F8-/- or PROC+/+;F9-/- mice in a needle puncture-induced knee-joint bleeding model. Additionally, we found that HAPC1573 significantly improved the thrombin generation of PROC+/+;F8-/- mice but not F8-/- mice, indicating that HAPC1573 enhanced the coagulant activity of hemophilia mice by modulating human APC in vivo. We further documented that HAPC1573 inhibited the APC anticoagulant activity to improve the clotting time of human plasma deficient of FVIII, FIX, FXI, FVII, VWF, FV, or FX. These results demonstrate that selectively blocking the anticoagulant activity of human APC may be an effective therapeutic and/or prophylactic approach for bleeding disorders lacking FVIII, FIX, or other clotting factors.


Asunto(s)
Hemofilia A , Animales , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Coagulación Sanguínea , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemorragia , Hemostasis , Humanos , Ratones , Proteína C/farmacología , Proteína C/uso terapéutico
14.
Blood ; 139(12): 1892-1902, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-34890454

RESUMEN

Rebalancing the hemostatic system by targeting endogenous anticoagulant pathways, like the protein C (PC) system, is being tested as a means of improving hemostasis in patients with hemophilia. Recent intravital studies of hemostasis demonstrated that, in some vascular contexts, thrombin activity is sequestered in the extravascular compartment. These findings raise important questions about the context-dependent contribution of activated PC (APC) to the hemostatic response, because PC activation occurs on the surface of endothelial cells. We used a combination of pharmacologic, genetic, imaging, and computational approaches to examine the relationships among thrombin spatial distribution, PC activation, and APC anticoagulant function. We found that inhibition of APC activity, in mice either harboring the factor V Leiden mutation or infused with an APC-blocking antibody, significantly enhanced fibrin formation and platelet activation in a microvascular injury model, consistent with the role of APC as an anticoagulant. In contrast, inhibition of APC activity had no effect on hemostasis after penetrating injury of the mouse jugular vein. Computational studies showed that differences in blood velocity, injury size, and vessel geometry determine the localization of thrombin generation and, consequently, the extent of PC activation. Computational predictions were tested in vivo and showed that when thrombin generation occurred intravascularly, without penetration of the vessel wall, inhibition of APC significantly increased fibrin formation in the jugular vein. Together, these studies show the importance of thrombin spatial distribution in determining PC activation during hemostasis and thrombosis.


Asunto(s)
Hemostáticos , Trombosis , Animales , Anticoagulantes/farmacología , Células Endoteliales/metabolismo , Fibrina/metabolismo , Hemostasis , Humanos , Ratones , Proteína C/farmacología , Trombina/metabolismo , Trombosis/metabolismo
15.
J Exp Med ; 219(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34846535

RESUMEN

Subcortical white matter (WM) stroke accounts for 25% of all strokes and is the second leading cause of dementia. Despite such clinical importance, we still do not have an effective treatment for ischemic WM stroke, and the mechanisms of WM postischemic neuroprotection remain elusive. 3K3A-activated protein C (APC) is a signaling-selective analogue of endogenous blood protease APC that is currently in development as a neuroprotectant for ischemic stroke patients. Here, we show that 3K3A-APC protects WM tracts and oligodendrocytes from ischemic injury in the corpus callosum in middle-aged mice by activating protease-activated receptor 1 (PAR1) and PAR3. We show that PAR1 and PAR3 were also required for 3K3A-APC's suppression of post-WM stroke microglia and astrocyte responses and overall improvement in neuropathologic and functional outcomes. Our data provide new insights into the neuroprotective APC pathway in the WM and illustrate 3K3A-APC's potential for treating WM stroke in humans, possibly including multiple WM strokes that result in vascular dementia.


Asunto(s)
Cuerpo Calloso/metabolismo , Isquemia/metabolismo , Oligodendroglía/metabolismo , Proteína C/metabolismo , Sustancia Blanca/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Cuerpo Calloso/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacología , Humanos , Isquemia/fisiopatología , Isquemia/prevención & control , Masculino , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Proteína C/farmacología , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/prevención & control
16.
Nutr Cancer ; 74(1): 278-287, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33533291

RESUMEN

Thymoquinone (TQ) combined with Cisplatin may augment its anticancer effect on hepatocellular carcinoma (HCC), through oxidative stress mitigation and endoplasmic reticulum (ER) protein modulation. Fifty adult male Wistar albino rats were assigned into five equal experimental groups (n = 10); 1) Control, 2) diethylnitrosamine/carbon tetrachloride-induced liver tumorigenesis model (HCC), 3) Cisplatin (2 mg.kg-1ip) treated rats, 4) Thymoquinone treated group (20 mg.kg-1oral), and 5) group treated with both drugs as in Groups 3 and 4. Treatment regimens started following model confirmation and continued for 4 weeks. In the HCC model, we detected elevated ER chaperone glucose-regulated protein-78 (GRP78) and reduced C/EBP-homologous protein (CHOP)-mediated apoptosis that was accompanied by the elevated alpha-fetoprotein (AFP) marker and deteriorated liver functions. Our original results indicated that Thymoquinone potentiated the pro-apoptotic effect of cisplatin by modulating GRP78/CHOP signaling. Cisplatin/TQ reduced the elevated GRP78 and induced CHOP-mediated apoptosis in the diseased liver tissues compared to the HCC and Cisplatin treated groups. Cisplatin/TQ combination normalized AFP levels and improved liver functions compared to both HCC and cisplatin groups alone. In conclusion, Thymoquinone enhanced the efficacy of Cisplatin in HCC treatment by modulating the GRP78/CHOP/caspase-3 pathway. Thymoquinone is recommended to achieve greater therapeutic benefits and reduce the cisplatin hepatotoxicity in HCC management.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Apoptosis , Benzoquinonas , Carcinogénesis , Carcinoma Hepatocelular/patología , Cisplatino/farmacología , Estrés del Retículo Endoplásmico , Glucosa/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Estrés Oxidativo , Proteína C/metabolismo , Proteína C/farmacología , Ratas , Ratas Wistar
17.
Front Immunol ; 12: 650184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868288

RESUMEN

Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection and is associated with high morbidity and mortality. Circulating histones (CHs), a group of damage-associated molecular pattern molecules mainly derived from neutrophil extracellular traps, play a crucial role in sepsis by mediating inflammation response, organ injury and death through Toll-like receptors or inflammasome pathways. Herein, we first elucidate the molecular mechanisms of histone-induced inflammation amplification, endothelium injury and cascade coagulation activation, and discuss the close correlation between elevated level of CHs and disease severity as well as mortality in patients with sepsis. Furthermore, current state-of-the-art on anti-histone therapy with antibodies, histone-binding proteins (namely recombinant thrombomodulin and activated protein C), and heparin is summarized to propose promising approaches for sepsis treatment.


Asunto(s)
Alarminas/sangre , Antiinflamatorios/farmacología , Histonas/sangre , Sepsis/diagnóstico , Alarminas/antagonistas & inhibidores , Alarminas/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Biomarcadores/sangre , Biomarcadores/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/inmunología , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Heparina/farmacología , Heparina/uso terapéutico , Histonas/antagonistas & inhibidores , Histonas/metabolismo , Humanos , Terapia Molecular Dirigida/métodos , Pronóstico , Proteína C/farmacología , Proteína C/uso terapéutico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Sepsis/sangre , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal/inmunología , Trombomodulina/uso terapéutico
18.
Thromb Haemost ; 121(11): 1448-1463, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33706396

RESUMEN

Activated protein C (APC) is an anticoagulant plasma serine protease which exhibits potent cytoprotective and anti-inflammatory activities. Here, we studied protective effects of APC on the proinflammatory function of polyinosinic:polycytidylic acid [poly(I:C)], a synthetic analog of viral double-stranded RNA, in cellular and animal models. Poly(I:C) induced histone H3 extranuclear translocation via interaction with toll-like receptor 3 in two established endothelial cell lines. Furthermore, poly(I:C) induced histone H3 extranuclear translocation in J774A.1 macrophages and human neutrophils and formation of macrophage and neutrophil extracellular traps (ETs). Mechanistically, poly(I:C) was found to upregulate expression of peptidylarginine deiminase 4 and enhance its interaction with histone H3, thereby leading to increased histone citrullination and neutrophil ET formation. Poly(I:C) elicited proinflammatory signaling responses by inducing nuclear factor kappa B activation and disrupting endothelial cell permeability. In vivo, poly(I:C) enhanced cell surface expression of Mac-1 on neutrophils in mice and facilitated their infiltration to lung tissues. Poly(I:C) also downregulated thrombomodulin expression in mouse tissues and reduced its circulating soluble level in plasma. We demonstrate in this study that APC and a signaling-selective mutant of APC effectively inhibit proinflammatory signaling effects of poly(I:C) in both cellular and animal models. We further demonstrate that unlike the requirement for endothelial protein C receptor on endothelial cells, the integrin Mac-1 is involved in the protease-activated receptor 1-dependent APC inhibition of macrophage ET formation in J774A.1 cells. Taken together, these results support a key role for APC signaling in inhibiting the viral mimetic-induced proinflammatory signaling responses and histone translocation-associated formation of ETs by innate immune cells.


Asunto(s)
Antiinflamatorios/farmacología , Células Endoteliales/efectos de los fármacos , Inflamación/prevención & control , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Proteína C/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Activación Enzimática , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Histonas/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Poli I-C , Proteína C/genética , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal
19.
Circ Res ; 128(4): 513-529, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33353373

RESUMEN

RATIONALE: While thrombin is the key protease in thrombus formation, other coagulation proteases, such as fXa (factor Xa) or aPC (activated protein C), independently modulate intracellular signaling via partially distinct receptors. OBJECTIVES: To study the differential effects of fXa or fIIa (factor IIa) inhibition on gene expression and inflammation in myocardial ischemia-reperfusion injury. METHODS AND RESULTS: Mice were treated with a direct fIIa inhibitor (fIIai) or direct fXa inhibitor (fXai) at doses that induced comparable anticoagulant effects ex vivo and in vivo (tail-bleeding assay and FeCl3-induced thrombosis). Myocardial ischemia-reperfusion injury was induced via left anterior descending ligation. We determined infarct size and in vivo aPC generation, analyzed gene expression by RNA sequencing, and performed immunoblotting and ELISA. The signaling-only 3K3A-aPC variant and inhibitory antibodies that blocked all or only the anticoagulant function of aPC were used to determine the role of aPC. Doses of fIIai and fXai that induced comparable anticoagulant effects resulted in a comparable reduction in infarct size. However, unbiased gene expression analyses revealed marked differences, including pathways related to sterile inflammation and inflammasome regulation. fXai but not fIIai inhibited sterile inflammation by reducing the expression of proinflammatory cytokines (IL [interleukin]-1ß, IL-6, and TNFα [tumor necrosis factor alpha]), as well as NF-κB (nuclear factor kappa B) and inflammasome activation. This anti-inflammatory effect was associated with reduced myocardial fibrosis 28 days post-myocardial ischemia-reperfusion injury. Mechanistically, in vivo aPC generation was higher with fXai than with fIIai. Inhibition of the anticoagulant and signaling properties of aPC abolished the anti-inflammatory effect associated with fXai, while inhibiting only the anticoagulant function of aPC had no effect. Combining 3K3A-aPC with fIIai reduced the inflammatory response, mimicking the fXai-associated effect. CONCLUSIONS: We showed that specific inhibition of coagulation via direct oral anticoagulants had differential effects on gene expression and inflammation, despite comparable anticoagulant effects and infarct sizes. Targeting individual coagulation proteases induces specific cellular responses unrelated to their anticoagulant effect.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inhibidores del Factor Xa/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Proteína C/uso terapéutico , Animales , Antiinflamatorios/farmacología , Inhibidores del Factor Xa/farmacología , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Proteína C/farmacología
20.
Stroke ; 51(7): 2236-2239, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32568648

RESUMEN

BACKGROUND AND PURPOSE: A recombinant engineered variant of APC (activated protein C), 3K3A-APC, lacks anticoagulant properties (<10%) while preserving APCs anti-inflammatory, anti-apoptotic, and neuroprotective functions and is very promising in clinical trials for ischemic stroke. Therapeutic intervention with single bolus administration of the third-generation tPA (tissue-type plasminogen activator), tenecteplase, is anticipated to be widely adopted for treatment of acute ischemic stroke. 3K3A-APC is well-tolerated in stroke patients dosed with alteplase, and in vitro studies show 3K3A-APC does not interfere with alteplase-induced clot lysis. The purpose of this in vitro study was to assess the influence of 3K3A-APC on tenecteplase-induced clot lysis. METHODS: Tenecteplase-mediated lysis of thrombin generated plasma clots of human normal pooled plasma was monitored in the presence of varying doses of 3K3A-APC. The effects on fibrinolysis by tenecteplase and alteplase were compared. RESULTS: The presence of 3K3A-APC shortened the time for clot lysis induced by tenecteplase at very low levels but not at higher therapeutic concentrations of tenecteplase. Comparisons of alteplase-mediated clot lysis to tenecteplase clot lysis showed that both thrombolytic agents behaved similarly in the presence of 3K3A-APC. CONCLUSIONS: These results indicate that 3K3A-APC does not interfere with tenecteplase's clot lysis function.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Fibrinolíticos/farmacología , Proteína C/farmacología , Proteínas Recombinantes/farmacología , Tenecteplasa/farmacología , Humanos , Técnicas In Vitro , Trombosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...