Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38860847

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by vasoconstriction and remodeling of small pulmonary arteries (PAs). Central to the remodeling process is a switch of pulmonary vascular cells to a proliferative, apoptosis-resistant phenotype. Plasminogen activator inhibitor-1 (PAI-1) is the primary physiological inhibitor of urokinase-type and tissue-type plasminogen activators (uPA and tPA), but its role in PAH is unsettled. Here, we report that: (1) PAI-1 is deficient in remodeled small PAs and in early-passage PA smooth muscle and endothelial cells (PASMCs and PAECs) from subjects with PAH compared to controls; (2) PAI-1-/- mice spontaneously develop pulmonary vascular remodeling associated with up-regulation of mTORC1 signaling, pulmonary hypertension (PH), and right ventricle (RV) hypertrophy; and (3) pharmacological inhibition of uPA in human PAH PASMCs suppresses pro-proliferative mTORC1 and SMAD3 signaling, restores PAI-1 levels, reduces proliferation and induces apoptosis in vitro, and prevents the development of SU5416/hypoxia-induced PH and RV hypertrophy in vivo in mice. These data strongly suggest that down-regulation of PAI-1 in small PAs promotes vascular remodeling and PH due to unopposed activation of uPA and consequent up-regulation of mTOR and TGF-b signaling in PASMCs, and call for further studies to determine the potential benefits of targeting the PAI-1/uPA imbalance to attenuate and/or reverse pulmonary vascular remodeling and PH.

2.
Blood ; 135(23): 2085-2093, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32187355

RESUMEN

Thromboembolism complicates disorders caused by immunoglobulin G (IgG)-containing immune complexes (ICs), but the underlying mechanisms are incompletely understood. Prior evidence indicates that induction of tissue factor (TF) on monocytes, a pivotal step in the initiation, localization, and propagation of coagulation by ICs, is mediated through Fcγ receptor IIa (FcγRIIa); however, the involvement of other receptors has not been investigated in detail. The neonatal Fc receptor (FcRn) that mediates IgG and albumin recycling also participates in cellular responses to IgG-containing ICs. Here we asked whether FcRn is also involved in the induction of TF-dependent factor Xa (FXa) activity by IgG-containing ICs by THP-1 monocytic cells and human monocytes. Induction of FXa activity by ICs containing IgG antibodies to platelet factor 4 (PF4) involved in heparin-induced thrombocytopenia (HIT), ß-2-glycoprotein-1 implicated in antiphospholipid syndrome, or red blood cells coated with anti-(α)-Rh(D) antibodies that mediate hemolysis in vivo was inhibited by a humanized monoclonal antibody (mAb) that blocks IgG binding to human FcRn. IgG-containing ICs that bind to FcγR and FcRn induced FXa activity, whereas IgG-containing ICs with an Fc engineered to be unable to engage FcRn did not. Infusion of an α-FcRn mAb prevented fibrin deposition after microvascular injury in a murine model of HIT in which human FcγRIIa was expressed as a transgene. These data implicate FcRn in TF-dependent FXa activity induced by soluble and cell-associated IgG-containing ICs. Antibodies to FcRn, now in clinical trials in warm autoimmune hemolytic anemia to lower IgG antibodies and IgG containing ICs may also reduce the risk of venous thromboembolism.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Heparina/toxicidad , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo , Trombocitopenia/inmunología , Tromboplastina/metabolismo , Animales , Anticoagulantes/toxicidad , Complejo Antígeno-Anticuerpo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Masculino , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Receptores Fc/genética , Receptores Fc/inmunología , Trombocitopenia/inducido químicamente , Trombocitopenia/metabolismo , Trombocitopenia/patología
3.
Molecules ; 27(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35408677

RESUMEN

Analytical control of protein and amino acid (AA) contents of animal tissues is an important problem in the fundamental and applied aspects. The aims of the work were the following: to measure the pig blood AAs; and to establish the correlations between AAs and biochemical parameters in dependence on the pig fattening duration. All 80 animals were divided onto 4 animal groups: 65, 72, 82, and 90 fattening days. The correlations between AAs and the total protein or its fractions (TP&F), nitrogen metabolites, carbohydrates, lipids, some enzymes in the pig blood for each of these animal groups obtained for the first time. The authors established the following total amounts of correlation coefficients (with reasonable p-values) in each of the group separately: group 1, 1* (p < 0.05); group 2, 0; group 3, 28* (p < 0.05) and 9** (p < 0.01); group 4, 28* (p < 0.05) and 25** (p < 0.01). Thus, about 82−90 days (groups 3 and 4) can be the optimal for the pig fattening, based on the correlation analysis for the numerous data of major AA and biochemical parameters of pig blood. These results can be useful for animal health monitoring and husbandry.


Asunto(s)
Crianza de Animales Domésticos , Enfermedades de los Porcinos , Aminoácidos , Crianza de Animales Domésticos/métodos , Animales , Nitrógeno , Porcinos
4.
J Biol Chem ; 292(50): 20528-20543, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28972182

RESUMEN

Lymphangioleiomyomatosis (LAM) is a fatal lung disease associated with germline or somatic inactivating mutations in tuberous sclerosis complex genes (TSC1 or TSC2). LAM is characterized by neoplastic growth of smooth muscle-α-actin-positive cells that destroy lung parenchyma and by the formation of benign renal neoplasms called angiolipomas. The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin slows progression of these diseases but is not curative and associated with notable toxicity at clinically effective doses, highlighting the need for better understanding LAM's molecular etiology. We report here that LAM lesions and angiomyolipomas overexpress urokinase-type plasminogen activator (uPA). Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts expressed higher uPA levels than their WT counterparts, resulting from the TSC inactivation. Inhibition of uPA expression in Tsc2-null cells reduced the growth and invasiveness and increased susceptibility to apoptosis. However, rapamycin further increased uPA expression in TSC2-null tumor cells and immortalized TSC2-null angiomyolipoma cells, but not in cells with intact TSC. Induction of glucocorticoid receptor signaling or forkhead box (FOXO) 1/3 inhibition abolished the rapamycin-induced uPA expression in TSC-compromised cells. Moreover, rapamycin-enhanced migration of TSC2-null cells was inhibited by the uPA inhibitor UK122, dexamethasone, and a FOXO inhibitor. uPA-knock-out mice developed fewer and smaller TSC2-null lung tumors, and introduction of uPA shRNA in tumor cells or amiloride-induced uPA inhibition reduced tumorigenesis in vivo These findings suggest that interference with the uPA-dependent pathway, when used along with rapamycin, might attenuate LAM progression and potentially other TSC-related disorders.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Linfangioleiomiomatosis/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Angiomiolipoma/tratamiento farmacológico , Angiomiolipoma/genética , Angiomiolipoma/metabolismo , Angiomiolipoma/patología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Interferencia de ARN , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Carga Tumoral/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/genética
5.
FASEB J ; 31(2): 761-770, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27836986

RESUMEN

Endothelial thrombomodulin (TM) regulates coagulation and inflammation via several mechanisms, including production of activated protein C (APC). Recombinant APC and soluble fragments of TM (sTM) have been tested in settings associated with insufficiency of the endogenous TM/APC pathway, such as sepsis. We previously designed a fusion protein of TM [single-chain variable fragment antibody (scFv)/TM] targeted to red blood cells (RBCs) to improve pharmacokinetics and antithrombotic effects without increasing bleeding. Here, scFv/TM was studied in mouse models of systemic inflammation and ischemia-reperfusion injury. Injected concomitantly with or before endotoxin, scFv/TM provided more potent protection against liver injury and release of pathological mediators than sTM, showing similar efficacy at up to 50-fold lower doses. scFv/TM provided protection when injected after endotoxin, whereas sTM did not, and augmented APC production by thrombin ∼50-fold more than sTM. However, scFv/TM injected after endotoxin did not reduce thrombin/antithrombin complexes; nor did antibodies that block APC anticoagulant activity suppress the prophylactic anti-inflammatory effect of scFv/TM. Therefore, similar to endogenous TM, RBC-anchored scFv/TM activates several protective pathways. Finally, scFv/TM was more effective at reducing cerebral infarct volume and alleviated neurological deficits than sTM after cerebral ischemia/reperfusion injury. These results indicate that RBC-targeted scFv/TM exerts multifaceted cytoprotective effects and may find utility in systemic and focal inflammatory and ischemic disorders.-Carnemolla, R., Villa, C. H., Greineder, C. F., Zaitseva, S., Patel, K. R., Kowalska, M. A., Atochin, D. N., Cines, D. B., Siegel, D. L., Esmon, C. T., Muzykantov, V. R. Targeting thrombomodulin to circulating red blood cells augments its protective effects in models of endotoxemia and ischemia-reperfusion injury.


Asunto(s)
Endotoxemia/prevención & control , Eritrocitos/metabolismo , Daño por Reperfusión/prevención & control , Trombomodulina/administración & dosificación , Trombomodulina/uso terapéutico , Animales , Inflamación/tratamiento farmacológico , Masculino , Proteínas de la Fusión de la Membrana , Ratones , Ratones Endogámicos C57BL , Trombomodulina/química
6.
J Biol Chem ; 291(29): 15029-45, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27151212

RESUMEN

Urokinase-type plasminogen activator (uPA) regulates angiogenesis and vascular permeability through proteolytic degradation of extracellular matrix and intracellular signaling initiated upon its binding to uPAR/CD87 and other cell surface receptors. Here, we describe an additional mechanism by which uPA regulates angiogenesis. Ex vivo VEGF-induced vascular sprouting from Matrigel-embedded aortic rings isolated from uPA knock-out (uPA(-/-)) mice was impaired compared with vessels emanating from wild-type mice. Endothelial cells isolated from uPA(-/-) mice show less proliferation and migration in response to VEGF than their wild type counterparts or uPA(-/-) endothelial cells in which expression of wild type uPA had been restored. We reported previously that uPA is transported from cell surface receptors to nuclei through a mechanism that requires its kringle domain. Intranuclear uPA modulates gene transcription by binding to a subset of transcription factors. Here we report that wild type single-chain uPA, but not uPA variants incapable of nuclear transport, increases the expression of cell surface VEGF receptor 1 (VEGFR1) and VEGF receptor 2 (VEGFR2) by translocating to the nuclei of ECs. Intranuclear single-chain uPA binds directly to and interferes with the function of the transcription factor hematopoietically expressed homeodomain protein or proline-rich homeodomain protein (HHEX/PRH), which thereby lose their physiologic capacity to repress the activity of vehgr1 and vegfr2 gene promoters. These studies identify uPA-dependent de-repression of vegfr1 and vegfr2 gene transcription through binding to HHEX/PRH as a novel mechanism by which uPA mediates the pro-angiogenic effects of VEGF and identifies a potential new target for control of pathologic angiogenesis.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Neovascularización Fisiológica , Factores de Transcripción/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Células K562 , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
7.
Invest New Drugs ; 34(4): 522-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27221730

RESUMEN

We have previously shown that the insulinotropic imidazoline compound RX871024 induces death of insulinoma MIN6 cells, an effect involving stimulation of c-Jun N-terminal kinase (JNK) and caspase 3. It has also been reported that AMP-activated protein kinase (AMPK) activates JNK and induces ß-cell death. Here we show that RX871024, but not another insulinotropic imidazoline compound (BL11282), suppressed AMPK activity in MIN6 cells. The inhibitory effect of RX871024 on AMPK was supported by the observation that the imidazoline induced lipid droplet formation in the cytoplasm of MIN6 cells. This reflects stimulation of anabolic pathways and inhibition of catabolic pathways in the cell that happen under conditions when AMPK is inhibited. Activation of AMPK by 5-aminoimidazole-4-carboxamide riboside (AICAR) elevated basal and cytokine-induced death in primary ß-cells and in insulinoma MIN6 cells. RX871024 aggravated AICAR-induced insulinoma MIN6 cell death regardless of the presence of pro-inflammatory cytokines. The specific cytotoxic effect of imidazoline compound RX871024 on insulinoma cell death but not primary ß-cell death is independent of its action on AMPK and may suggest the possibility of using this type of compound in the treatment of insulinomas.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Indoles/farmacología , Insulinoma/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/metabolismo , Ratones Obesos , Fosforilación/efectos de los fármacos , Ribonucleósidos/farmacología
8.
FASEB J ; 29(8): 3483-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25953848

RESUMEN

Anchoring pharmacologic agents to the vascular lumen has the potential to modulate critical processes at the blood-tissue interface, avoiding many of the off-target effects of systemically circulating agents. We report a novel strategy for endothelial dual targeting of therapeutics, which both enhances drug delivery and enables targeted agents to partner enzymatically to generate enhanced biologic effect. Based on the recent discovery that paired antibodies directed to adjacent epitopes of platelet endothelial cell adhesion molecule (PECAM)-1 stimulate each other's binding, we fused single-chain fragments (scFv) of paired anti-mouse PECAM-1 antibodies to recombinant murine thrombomodulin (TM) and endothelial protein C receptor (EPCR), endothelial membrane proteins that partner in activation of protein C (PC). scFv/TM and scFv/EPCR bound to mouse endothelial PECAM-1 with high affinity (EC50 1.5 and 3.8 nM, respectively), and codelivery induced a 5-fold increase in PC activation not seen when TM and EPCR are anchored to distinct cell adhesion molecules. In a mouse model of acute lung injury, dual targeting reduces both the expression of lung inflammatory markers and trans-endothelial protein leak by as much as 40%, as compared to either agent alone. These findings provide proof of principle for endothelial dual targeting, an approach with numerous potential biomedical applications.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Epítopos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/metabolismo , Trombomodulina/metabolismo
9.
Am J Physiol Endocrinol Metab ; 309(4): E418-27, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26126683

RESUMEN

Inflammation and impaired mitochondrial oxidative phosphorylation are considered key players in the development of several metabolic disorders, including diabetes. We have previously shown inflammation and mitochondrial dysfunction in the hypothalamus of an animal model for anorexia, the anx/anx mouse. Moreover, increased incidence of eating disorders, e.g., anorexia nervosa, has been observed in diabetic individuals. In the present investigation we evaluated whether impaired mitochondrial phosphorylation and inflammation also occur in endocrine pancreas of anorectic mice, and if glucose homeostasis is disturbed. We show that anx/anx mice exhibit marked glucose intolerance associated with reduced insulin release following an intraperitoneal injection of glucose. In contrast, insulin release from isolated anx/anx islets is increased after stimulation with glucose or KCl. In isolated anx/anx islets there is a strong downregulation of the mitochondrial complex I (CI) assembly factor, NADH dehydrogenase (ubiquinone) 1α subcomplex, assembly factor 1 (Ndufaf1), and a reduced CI activity. In addition, we show elevated concentrations of free fatty acids (FFAs) in anx/anx serum and increased macrophage infiltration (indicative of inflammation) in anx/anx islets. However, isolated islets from anx/anx mice cultured in the absence of FFAs do not exhibit increased inflammation. We conclude that the phenotype of the endocrine pancreas of the anx/anx mouse is characterized by increased levels of circulating FFAs, as well as inflammation, which can inhibit insulin secretion in vivo. The anx/anx mouse may represent a useful tool for studying molecular mechanisms underlying the association between diabetes and eating disorders.


Asunto(s)
Anorexia/fisiopatología , Intolerancia a la Glucosa/fisiopatología , Células Secretoras de Insulina/fisiología , Animales , Anorexia/complicaciones , Anorexia/metabolismo , Anorexia/patología , Glucemia/metabolismo , Recuento de Células , Células Cultivadas , Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de los Órganos , Páncreas/patología
10.
Blood ; 119(20): 4779-85, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22493296

RESUMEN

Thrombin generates fibrin and activates platelets and endothelium, causing thrombosis and inflammation. Endothelial thrombomodulin (TM) changes thrombin's substrate specificity toward cleavage of plasma protein C into activated protein C (APC), which opposes its thrombotic and inflammatory activities. Endogenous TM activity is suppressed in pathologic conditions, and antithrombotic interventions involving soluble TM are limited by rapid blood clearance. To overcome this problem, we fused TM with a single chain fragment (scFv) of an antibody targeted to red blood cells. scFv/TM catalyzes thrombin-mediated generation of activated protein C and binds to circulating RBCs without apparent damage, thereby prolonging its circulation time and bioavailability orders of magnitude compared with soluble TM. In animal models, a single dose of scFv/TM, but not soluble TM, prevents platelet activation and vascular occlusion by clots. Thus, scFv/TM serves as a prodrug and provides thromboprophylaxis at low doses (0.15 mg/kg) via multifaceted mechanisms inhibiting platelets and coagulation.


Asunto(s)
Quimioprevención/métodos , Sistemas de Liberación de Medicamentos/métodos , Eritrocitos/efectos de los fármacos , Trombomodulina/administración & dosificación , Trombosis/prevención & control , Animales , Células Cultivadas , Drosophila , Eritrocitos/metabolismo , Eritrocitos/fisiología , Humanos , Ratones , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Unión Proteica , Proteína C/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/metabolismo , Anticuerpos de Cadena Única/uso terapéutico , Trombomodulina/química , Trombomodulina/metabolismo , Trombomodulina/uso terapéutico
11.
Biopolymers ; 102(3): 252-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24549714

RESUMEN

Glucagon-like peptide 1 (GLP-1) and related peptide agonists have been extensively investigated for glycaemic control in Type 2 diabetes, and may also have therapeutic applications for other diseases. Due to the short half-life (t1/2 < 2 min) of the endogenous peptide, caused by proteolytic degradation and renal clearance, different strategies for half-life extension and sustained release have been explored. In the present study, conjugates between a GLP-1 analogue and a 5 kDa albumin-binding domain (ABD) derived from streptococcal protein G have been chemically synthesized and evaluated. ABD binds with high affinity to human serum albumin, which is highly abundant in plasma and functions as a drug carrier in the circulation. Three different GLP-1-ABD conjugates, with the two peptides connected by linkers of two, four, and six PEG units, respectively, were synthesized and tested in mouse pancreatic islets at high (11 mM) and low (3 mM) glucose concentration. Insulin release upon stimulation was shown to be glucose-dependent, showing no significant difference between the three different GLP-1-ABD conjugates and unconjugated GLP-1 analogue. The biological activity, in combination with the high affinity binding to albumin, make the GLP-1-ABD conjugates promising GLP-1 receptor agonists expected to show extended in vivo half-life.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/química , Albúmina Sérica/química , Animales , Técnicas Biosensibles , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Semivida , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos , Ratones Obesos , Péptidos/síntesis química , Péptidos/química , Estructura Terciaria de Proteína , Ratas
12.
ScientificWorldJournal ; 2014: 285405, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25383365

RESUMEN

Conjugates of the photoactivated rhodamine dyes with biopolymers (proteins, polysaccharides, and nucleic acids) are important tools for microscopic investigation of biological tissue. In this study, a precursor of the photoactivated fluorescent dye (PFD) has been successfully used for staining of numerous mammalian cells lines and for conjugate formation with chitosan ("Chitosan-PFD") and histone H1 ("Histone H1.3-PFD"). The intensive fluorescence has been observed after photoactivation of these conjugates inside cells (A431, HaCaT, HEK239, HBL-100, and MDCK). Developed procedures and obtained data are important for further application of novel precursors of fluorescent dyes ("caged" dyes) for microscopic probing of biological objects. Thus, the synthesized "Chitosan-PFD" and "Histone H1-PFD" have been successfully applied in this study for intracellular transport visualization by fluorescent microscopy.


Asunto(s)
Biopolímeros/química , Colorantes Fluorescentes/química , Rodaminas/síntesis química , Animales , Rastreo Celular , Colorantes Fluorescentes/síntesis química , Células HEK293 , Humanos , Microscopía Fluorescente , Proteínas/química , Rodaminas/química , Coloración y Etiquetado
13.
Blood Adv ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38805575

RESUMEN

Fibrinolytics delivered into the general circulation lack selectivity for nascent thrombi, reducing efficacy and increasing the risk of bleeding. Urokinase-type plasminogen activator (uPA) transgenically expressed within murine platelets provided targeted thromboprophylaxis without causing bleeding, but is clinically infeasible. Recent advances in generating megakaryocytes prompted us to develop a potentially clinically relevant means to produce "anti-thrombotic" platelets from CD34+ hematopoietic stem cell-derived in vitro-grown megakaryocytes. CD34+-megakaryocytes internalize and store in -granules single-chain uPA (scuPA) and a plasmin-resistant thrombin-activatable variant (uPAT). Both uPAs co-localized with internalized factor V (FV), fibrinogen and plasminogen, low-density lipoprotein receptor-related protein 1 (LRP1), and interferon-induced transmembrane protein 3, but not with endogenous von Willebrand factor (VWF). Endocytosis of uPA by CD34+-megakaryocytes was mediated, in part, via LRP1 and IIb3. scuPA-containing megakaryocytes degraded endocytosed intragranular FV, but not endogenous VWF in the presence of internalized plasminogen, whereas uPAT-megakaryocytes did not significantly degrade either protein. We used a carotid-artery injury model in NOD-scid IL2rnull (NSG) mice homozygous for VWFR1326H (a mutation switching binding VWF specificity from mouse to human glycoprotein Ib) to test whether platelets derived from scuPA- or uPAT-megakaryocytes would prevent thrombus formation. NSG/VWFR1326H mice exhibited a lower thrombotic burden after carotid artery injury compared to NSG mice unless infused with human platelets or megakaryocytes, whereas intravenous injection of uPA-megakaryocytes generated sufficient uPA-containing human platelets to lyse nascent thrombi. These studies describe the use of in vitro-generated megakaryocytes as a potential platform for delivering uPA or other ectopic proteins within platelet -granules to sites of vascular injury.

14.
J Pharmacol Exp Ther ; 347(2): 339-45, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23965383

RESUMEN

Thrombomodulin (TM) is a glycoprotein normally present in the membrane of endothelial cells that binds thrombin and changes its substrate specificity to produce activated protein C (APC) that has antithrombotic and anti-inflammatory features. To compensate for loss of endogenous TM in pathology, we have fused recombinant TM with single chain variable fragment (scFv) of an antibody to mouse platelet endothelial cell adhesion molecule-1 (PECAM). This fusion, anti-PECAM scFv/TM, anchors on the endothelium, stimulates APC production, and provides therapeutic benefits superior to sTM in animal models of acute thrombosis and inflammation. However, in conditions of oxidative stress typical of vascular inflammation, TM is inactivated via oxidation of the methionine 388 (M388) residue. Capitalizing on the reports that M388L mutation renders TM resistant to oxidative inactivation, in this study we designed a mutant anti-PECAM scFv/TM M388L. This mutant has the same APC-producing capacity and binding to target cells, yet, in contrast to wild-type fusion, it retains APC-producing activity in an oxidizing environment in vitro and in vivo. Therefore, oxidant resistant mutant anti-PECAM scFv/TM M388L is a preferable targeted biotherapeutic to compensate for loss of antithrombotic and anti-inflammatory TM functions in the context of vascular oxidative stress.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/farmacología , Proteína C/biosíntesis , Proteínas Recombinantes de Fusión/farmacología , Trombomodulina/genética , Animales , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Tiempo de Tromboplastina Parcial , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética , Especificidad por Sustrato , Trombina/metabolismo
15.
Blood ; 117(18): 4964-7, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21389322

RESUMEN

Studies in animal models have shown that plasminogen activators bound to erythrocytes (RBC-PA) have an extended lifetime in the circulation and are safer than free PAs. RBC-PAs incorporate into nascent thrombi, which are focally lysed from within, an attractive thromboprophylactic option. In static systems, RBC-PAs cleave surrounding fibrin fibers, forming pores larger than the cells themselves, and move around the pore edges, enlarging them until eventual clot dissolution. We hypothesized that under flow in blood vessels, RBC-PAs form functional patent channels before clot dissolution. Here we used perfusion chambers to study clot lysis by RBC-PAs under static versus arterial and venous flow conditions. We found that flow decelerates bulk clot lysis but quickly generates patent channels filled with passing RBCs, via pore enlargement and merging in the direction of flow. Formation of such channels by RBC-PAs may help rescue ischemic tissue before bulk dissolution of potentially occlusive clots.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinolíticos/farmacología , Animales , Coagulación Sanguínea/fisiología , Eritrocitos/efectos de los fármacos , Eritrocitos/fisiología , Hemorreología , Humanos , Técnicas In Vitro , Ratones , Microscopía Confocal , Modelos Biológicos , Activadores Plasminogénicos/farmacología
16.
Animals (Basel) ; 13(5)2023 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-36899709

RESUMEN

Many of the micro- and macro-elements (MMEs) required by the body are found in environmental objects in concentrations different from their original concentration that can lead to dangerous animal diseases ("microelementoses"). The aim was to study the features of MME (accumulating in wild and exotic animals) in connection with particular diseases. The work using 67 mammal species from four Russian zoological institutions was completed in 2022. Studies of 820 cleaned and defatted samples (hair, fur, etc.) after "wet-acid-ashing" on an electric stove and in a muffle furnace were performed using a Kvant-2A atomic absorption spectrometer. The content of zinc, copper, iron, cadmium, lead, and arsenic was assessed. The level of MME accumulation in the animal body contributes not only to the MME status and the development of various concomitant diseases, but the condition itself can occur by intake of a number of micronutrients and/or drugs. Particular correlations between the accumulation of Zn and skin, oncological diseases, Cu-musculoskeletal, cardiovascular diseases, Fe-oncological diseases, Pb-metabolic, nervous, oncological diseases, and Cd-cardiovascular diseases were established. Therefore, monitoring of the MME status of the organism must be carried out regularly (optimally once every 6 months).

17.
Animals (Basel) ; 13(10)2023 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-37238136

RESUMEN

Goats are ubiquitous, including in hot and dry regions, while also being very sensitive to climate fluctuations, expressed in temperature differences. This affects their productivity and milk quality. Adaptation to heat requires high energy costs, affects "neurohumoral" regulation and is accompanied by oxidative stress with the increased production of free radicals. The aim was to study the main biochemical parameters of goat milk and its antioxidant activity depending on the season of the year. Sampling was carried out in April, June, August and October. Analysis of the biochemical components and antioxidant activity of goat milk was performed using modern analytical systems. From spring to autumn, the mass fraction of true or crude proteins in goat milk increased by 14.6-63.7% or by 12.3-52.1%, and the mass fraction of caseins also increased by 13.6-60.6%. For vitamin C level and the total amount of water-soluble antioxidants, a pronounced gradual decrease from spring to autumn was observed. In the summer period, a small increase in the carotene level in milk (by 3.0-6.1% compared to April) was established. Vitamin A content increased by 86.5% (June) or by 70.3% (October) compared to April. Thus, the numerous significant changes in the major parameters of goat's milk depending on the season were revealed.

18.
bioRxiv ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38106191

RESUMEN

Our prior finding that uPA endogenously expressed and stored in the platelets of transgenic mice prevented thrombus formation without causing bleeding, prompted us to develop a potentially clinically relevant means of generating anti-thrombotic human platelets in vitro from CD34 + hematopoietic cell-derived megakaryocytes. CD34 + -megakaryocytes internalize and store in α-granules single-chain uPA (scuPA) and a uPA variant modified to be plasmin-resistant, but thrombin-activatable, (uPAT). Both uPAs co-localized with internalized factor V (FV), fibrinogen and plasminogen, low-density lipoprotein receptor-related protein 1 (LRP1), and interferon-induced transmembrane protein 3 (IFITM3), but not with endogenous von Willebrand factor (VWF). Endocytosis of uPA by CD34 + -\megakaryocytes was mediated in part via LRP1 and αIIbß3. scuPA-containing megakaryocytes degraded endocytosed intragranular FV, but not endogenous VWF, in the presence of internalized plasminogen, whereas uPAT-megakaryocytes did not significantly degrade either protein. We used a carotid-artery injury model in NOD-scid IL2rγnull (NSG) mice homozygous for VWF R1326H (a mutation switching binding VWF specificity from mouse to human glycoprotein IbmlIX) to test whether platelets derived from scuPA-MKs or uPAT-Mks would prevent thrombus formation. NSG/VWF R1326H mice exhibited a lower thrombotic burden after carotid artery injury compared to NSG mice unless infused with human platelets or MKs, whereas intravenous injection of either uPA-containing megakaryocytes into NSG/VWF R1326H generated sufficient uPA-containing human platelets to lyse nascent thrombi. These studies suggest the potential to deliver uPA or potentially other ectopic proteins within platelet α-granules from in vitro- generated megakaryocytes. Key points: Unlike platelets, in vitro-grown megakaryocytes can store exogenous uPA in its α-granules.uPA uptake involves LRP1 and αIIbß3 receptors and is functionally available from activated platelets.

19.
J Thromb Haemost ; 21(3): 652-666, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36696211

RESUMEN

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a serious thrombotic disorder caused by ultralarge immune complexes (ULICs) containing platelet factor 4 (PF4) and heparin that form the HIT antigen, together with a subset of anti-PF4 antibodies. ULICs initiate prothrombotic responses by engaging Fcγ receptors on platelets, neutrophils, and monocytes. Contemporary anti-thrombotic therapy for HIT is neither entirely safe nor entirely successful and acts downstream of ULIC formation and Fcγ receptor-initiated generation of thrombin. OBJECTIVES: To determine whether HIT antigen and ULIC formation and stability could be modified favorably by inhibiting PF4-heparin interactions with fondaparinux, together with blocking formation of PF4 tetramers using a humanized monoclonal anti-PF4 antibody (hRTO). METHODS: Results: The combination of fondaparinux and hRTO inhibited HIT antigen formation, promoted antigen dissociation, inhibited ULIC formation, and promoted ULIC disassembly at concentrations below the effective concentration of either alone and blocked Fcγ receptor-dependent induction of factor Xa activity by monocytic THP1 cells and activation of human platelets in whole blood. Combined with hRTO, fondaparinux inhibited HIT antigen and immune complex formation and activation through Fcγ receptors at concentrations at or below those used clinically to inhibit FXa coagulant activity. CONCLUSIONS: HIT antigen and immune complexes are dynamic and amenable to modulation. Fondaparinux can be converted from an anticoagulant that acts at a downstream amplification step into a rationale, disease-specific intervention that blocks ULIC formation. Interventions that prevent ULIC formation and stability might increase the efficacy, permit use of lower doses, shorten the duration of antithrombotic therapy, and help prevent this serious thrombotic disorder.


Asunto(s)
Trombocitopenia , Trombosis , Humanos , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticoagulantes/efectos adversos , Complejo Antígeno-Anticuerpo , Fondaparinux/efectos adversos , Heparina/efectos adversos , Factor Plaquetario 4 , Receptores de IgG , Trombosis/etiología
20.
bioRxiv ; 2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37790328

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive and potentially a rapidly fatal disease characterized by vasoconstriction and remodeling of small pulmonary arteries (PA) leading to increased pulmonary vascular resistance and right heart failure. Central to the remodeling process is a switch of the smooth muscle cells in small PAs (PASMC) to a proliferative, apoptosis-resistant phenotype. There is reason to suspect that the plasminogen activator system may play an important role in the remodeling program in PAH based on its roles in vascular post-injury restenosis, fibrosis, angiogenesis and tumorigenesis. Plasminogen activator inhibitor-1 (PAI-1) is the primary physiological inhibitor of the plasminogen activators - urokinase-type and tissue-type (uPA and tPA, respectively). Immunohisto- chemical and immunoblot analyses revealed that PAI-1 was deficient in smooth muscle areas of small remodeled PAs and early-passage PASMC from subjects with PAH compared to non-PAH controls. PAI1-/- male and female mice developed spontaneous pulmonary vascular remodeling and pulmonary hypertension (PH) as evidenced by significant increase in PA medial thickness, systolic right ventricular pressure, and right ventricular hypertrophy. Lastly, the uPA inhibitors upamostat (WX-671) and amiloride analog BB2-30F down-regulated mTORC1 and SMAD3, restored PAI-1 levels, reduced proliferation, and induced apoptosis in human PAH PASMC. We examined the effect of inhibition of uPA catalytic activity by BB2-30F on the development of SU5416/Hypoxia (SuHx)-induced PH in mice. Vehicletreated SuHx-exposed mice had up-regulated mTORC1 in small PAs, developed pulmonary vascular remodeling and PH, as evidenced by significant increase of PA MT, sRVP, RV hypertrophy, and a significant decrease in the pulmonary artery acceleration time/pulmonary ejection time (PAAT/PET) ratio compared to age- and sex-matched normoxia controls, whereas BB2-30F-treated group was protected from all these pathological changes. Taken together, our data strongly suggest that PAI-1 down- regulation in PASMC from human PAH lungs promotes PASMC hyper-proliferation, remodeling, and spontaneous PH due to unopposed uPA activation. Further studies are needed to determine the potential benefits of targeting the PAI-1/uPA imbalance to attenuate the progression and/or reverse pulmonary vascular remodeling and PH.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda