Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(1): e1010227, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35041705

RESUMEN

The blood-clotting protein fibrin(ogen) plays a critical role in host defense against invading pathogens, particularly against peritoneal infection by the Gram-positive microbe Staphylococcus aureus. Here, we tested the hypothesis that direct binding between fibrin(ogen) and S. aureus is a component of the primary host antimicrobial response mechanism and prevention of secondary microbe dissemination from the peritoneal cavity. To establish a model system, we showed that fibrinogen isolated from FibγΔ5 mice, which express a mutant form lacking the final 5 amino acids of the fibrinogen γ chain (termed fibrinogenγΔ5), did not support S. aureus adherence when immobilized and clumping when in suspension. In contrast, purified wildtype fibrinogen supported robust adhesion and clumping that was largely dependent on S. aureus expression of the receptor clumping factor A (ClfA). Following peritoneal infection with S. aureus USA300, FibγΔ5 mice displayed worse survival compared to WT mice coupled to reduced bacterial killing within the peritoneal cavity and increased dissemination of the microbes into circulation and distant organs. The failure of acute bacterial killing, but not enhanced dissemination, was partially recapitulated by mice infected with S. aureus USA300 lacking ClfA. Fibrin polymer formation and coagulation transglutaminase Factor XIII each contributed to killing of the microbes within the peritoneal cavity, but only elimination of polymer formation enhanced systemic dissemination. Host macrophage depletion or selective elimination of the fibrin(ogen) ß2-integrin binding motif both compromised local bacterial killing and enhanced S. aureus systemic dissemination, suggesting fibrin polymer formation in and of itself was not sufficient to retain S. aureus within the peritoneal cavity. Collectively, these findings suggest that following peritoneal infection, the binding of S. aureus to stabilized fibrin matrices promotes a local, macrophage-mediated antimicrobial response essential for prevention of microbe dissemination and downstream host mortality.


Asunto(s)
Fibrinógeno/inmunología , Peritonitis/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Coagulasa/inmunología , Coagulasa/metabolismo , Fibrina/metabolismo , Ratones , Peritonitis/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443167

RESUMEN

The blood-clotting protein fibrinogen has been implicated in host defense following Staphylococcus aureus infection, but precise mechanisms of host protection and pathogen clearance remain undefined. Peritonitis caused by staphylococci species is a complication for patients with cirrhosis, indwelling catheters, or undergoing peritoneal dialysis. Here, we sought to characterize possible mechanisms of fibrin(ogen)-mediated antimicrobial responses. Wild-type (WT) (Fib+) mice rapidly cleared S. aureus following intraperitoneal infection with elimination of ∼99% of an initial inoculum within 15 min. In contrast, fibrinogen-deficient (Fib-) mice failed to clear the microbe. The genotype-dependent disparity in early clearance resulted in a significant difference in host mortality whereby Fib+ mice uniformly survived whereas Fib- mice exhibited high mortality rates within 24 h. Fibrin(ogen)-mediated bacterial clearance was dependent on (pro)thrombin procoagulant function, supporting a suspected role for fibrin polymerization in this mechanism. Unexpectedly, the primary host initiator of coagulation, tissue factor, was found to be dispensable for this antimicrobial activity. Rather, the bacteria-derived prothrombin activator vWbp was identified as the source of the thrombin-generating potential underlying fibrin(ogen)-dependent bacterial clearance. Mice failed to eliminate S. aureus deficient in vWbp, but clearance of these same microbes in WT mice was restored if active thrombin was administered to the peritoneal cavity. These studies establish that the thrombin/fibrinogen axis is fundamental to host antimicrobial defense, offer a possible explanation for the clinical observation that coagulase-negative staphylococci are a highly prominent infectious agent in peritonitis, and suggest caution against anticoagulants in individuals susceptible to peritoneal infections.


Asunto(s)
Fibrinógeno/metabolismo , Peritonitis/metabolismo , Protrombina/metabolismo , Animales , Antibacterianos/metabolismo , Antiinfecciosos/metabolismo , Coagulación Sanguínea , Coagulasa/metabolismo , Femenino , Fibrina/metabolismo , Fibrinógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Tromboplastina
3.
PLoS Genet ; 15(3): e1007948, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30870413

RESUMEN

Glial cells regulate multiple aspects of synaptogenesis. In the absence of Schwann cells, a peripheral glial cell, motor neurons initially innervate muscle but then degenerate. Here, using a genetic approach, we show that neural activity-regulated negative factors produced by muscle drive neurodegeneration in Schwann cell-deficient mice. We find that thrombin, the hepatic serine protease central to the hemostatic coagulation cascade, is one such negative factor. Trancriptomic analysis shows that expression of the antithrombins serpin C1 and D1 is significantly reduced in Schwann cell-deficient mice. In the absence of peripheral neuromuscular activity, neurodegeneration is completely blocked, and expression of prothrombin in muscle is markedly reduced. In the absence of muscle-derived prothrombin, neurodegeneration is also markedly reduced. Together, these results suggest that Schwann cells regulate NMJs by opposing the effects of activity-regulated, muscle-derived negative factors and provide the first genetic evidence that thrombin plays a central role outside of the coagulation system.


Asunto(s)
Antitrombina III/genética , Cofactor II de Heparina/genética , Unión Neuromuscular/genética , Protrombina/genética , Sinapsis/genética , Animales , Perfilación de la Expresión Génica , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Músculo Esquelético/metabolismo , Degeneración Nerviosa/genética , Neuroglía , Unión Neuromuscular/crecimiento & desarrollo , Células de Schwann/metabolismo , Trombina/genética
4.
J Neurosci ; 37(14): 3776-3788, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28275164

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory, demyelinating disease of the CNS. Fibrinogen deposition at sites of blood-brain barrier breakdown is a prominent feature of neuroinflammatory disease and contributes to disease severity. Plasminogen, the primary fibrinolytic enzyme, also modifies inflammatory processes. We used a murine model of MS, experimental autoimmune encephalomyelitis (EAE), to evaluate the hypothesis that the loss of plasminogen would exacerbate neuroinflammatory disease. However, contrary to initial expectations, EAE-challenged plasminogen-deficient (Plg-) mice developed significantly delayed disease onset and reduced disease severity compared with wild-type (Plg+) mice. Similarly, pharmacologic inhibition of plasmin activation with tranexamic acid also delayed disease onset. The T-cell response to immunization was similar between genotypes, suggesting that the contribution of plasminogen was downstream of the T-cell response. Spinal cords from EAE-challenged Plg- mice demonstrated significantly decreased demyelination and microglial/macrophage accumulation compared with Plg+ mice. Although fibrinogen-deficient mice or mice with combined deficiencies of plasminogen and fibrinogen had decreased EAE severity, they did not exhibit the delay in EAE disease onset, as seen in mice with plasminogen deficiency alone. Together, these data suggest that plasminogen and plasmin-mediated fibrinolysis is a key modifier of the onset of neuroinflammatory demyelination.SIGNIFICANCE STATEMENT Multiple sclerosis is a severe, chronic, demyelinating disease. Understanding the pathobiology related to the autoreactive T-cell and microglial/macrophage demyelinating response is critical to effectively target therapeutics. We describe for the first time that deficiency of plasminogen, the key fibrinolytic enzyme, delays disease onset and protects from the development of the paralysis associated with a murine model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Administration of a widely used, pharmacologic inhibitor of plasminogen activation, tranexamic acid, also delays the onset of neuroinflammation associated with EAE.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/prevención & control , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Parálisis/metabolismo , Parálisis/prevención & control , Plasminógeno/deficiencia , Animales , Células Cultivadas , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parálisis/patología
5.
Blood ; 128(5): 721-31, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27252233

RESUMEN

Thrombin-mediated proteolysis is central to hemostatic function but also plays a prominent role in multiple disease processes. The proteolytic conversion of fII to α-thrombin (fIIa) by the prothrombinase complex occurs through 2 parallel pathways: (1) the inactive intermediate, prethrombin; or (2) the proteolytically active intermediate, meizothrombin (fIIa(MZ)). FIIa(MZ) has distinct catalytic properties relative to fIIa, including diminished fibrinogen cleavage and increased protein C activation. Thus, fII activation may differentially influence hemostasis and disease depending on the pathway of activation. To determine the in vivo physiologic and pathologic consequences of restricting thrombin generation to fIIa(MZ), mutations were introduced into the endogenous fII gene, resulting in expression of prothrombin carrying 3 amino acid substitutions (R157A, R268A, and K281A) to limit activation events to yield only fIIa(MZ) Homozygous fII(MZ) mice are viable, express fII levels comparable with fII(WT) mice, and have reproductive success. Although in vitro studies revealed delayed generation of fIIa(MZ) enzyme activity, platelet aggregation by fII(MZ) is similar to fII(WT) Consistent with prior analyses of human fIIa(MZ), significant prolongation of clotting times was observed for fII(MZ) plasma. Adult fII(MZ) animals displayed significantly compromised hemostasis in tail bleeding assays, but did not demonstrate overt bleeding. More notably, fII(MZ) mice had 2 significant phenotypic advantages over fII(WT) animals: protection from occlusive thrombosis after arterial injury and markedly diminished metastatic potential in a setting of experimental tumor metastasis to the lung. Thus, these novel animals will provide a valuable tool to assess the role of both fIIa and fIIa(MZ) in vivo.


Asunto(s)
Precursores Enzimáticos/metabolismo , Hemostasis , Protrombina/metabolismo , Trombina/metabolismo , Alelos , Animales , Coagulación Sanguínea , Retracción del Coagulo , Venenos de Crotálidos , Embrión de Mamíferos/metabolismo , Fibrosis , Metaloendopeptidasas , Ratones Endogámicos C57BL , Miocardio/patología , Metástasis de la Neoplasia , Agregación Plaquetaria , Análisis de Supervivencia , Trombosis/metabolismo , Trombosis/patología
6.
Blood ; 125(3): 427-37, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25336631

RESUMEN

Rheumatoid arthritis is a chronic inflammatory disease characterized by synovial hyperplasia, inflammatory cell infiltration, irreversible cartilage and bone destruction, and exuberant coagulation system activity within joint tissue. Here, we demonstrate that the coagulation transglutaminase, factor XIII (fXIII), drives arthritis pathogenesis by promoting local inflammatory and tissue degradative and remodeling events. All pathological features of collagen-induced arthritis (CIA) were significantly reduced in fXIII-deficient mice. However, the most striking difference in outcome was the preservation of cartilage and bone in fXIIIA(-/-) mice concurrent with reduced osteoclast numbers and activity. The local expression of osteoclast effectors receptor activator of nuclear factor-κB ligand (RANKL) and tartrate resistant acid phosphatase were significantly diminished in CIA-challenged and even unchallenged fXIIIA(-/-) mice relative to wild-type animals, but were similar in wild-type and fibrinogen-deficient mice. Impaired osteoclast formation in fXIIIA(-/-) mice was not due to an inherent deficiency of monocyte precursors, but it was linked to reduced RANKL-driven osteoclast formation. Furthermore, treatment of mice with the pan-transglutaminase inhibitor cystamine resulted in significantly diminished CIA pathology and local markers of osteoclastogenesis. Thus, eliminating fXIIIA limits inflammatory arthritis and protects from cartilage and bone destruction in part through mechanisms linked to reduced RANKL-mediated osteoclastogenesis. In summary, therapeutic strategies targeting fXIII activity may prove beneficial in limiting arthropathies and other degenerative bone diseases.


Asunto(s)
Artritis Experimental/patología , Enfermedades Óseas/fisiopatología , Factor XIII/fisiología , Inflamación/fisiopatología , Osteoclastos/patología , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Western Blotting , Enfermedades Óseas/complicaciones , Diferenciación Celular , Células Cultivadas , Colágeno/toxicidad , Femenino , Inflamación/complicaciones , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Osteoclastos/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Blood ; 126(15): 1844-55, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26286849

RESUMEN

Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Terapia Genética , Hipertensión Pulmonar/prevención & control , Inflamación/prevención & control , Protrombina/fisiología , Enfermedades Vasculares/prevención & control , Anemia de Células Falciformes/mortalidad , Anemia de Células Falciformes/fisiopatología , Animales , Coagulación Sanguínea , Células Cultivadas , Hipertensión Pulmonar/etiología , Técnicas para Inmunoenzimas , Inflamación/etiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Oligorribonucleótidos Antisentido/farmacología , Protrombina/antagonistas & inhibidores , Tasa de Supervivencia , Trombina/metabolismo , Enfermedades Vasculares/etiología
8.
Blood ; 126(17): 2047-58, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26228483

RESUMEN

Fibrin(ogen) is central to hemostasis and thrombosis and also contributes to multiple physiologic and pathologic processes beyond coagulation. However, the precise contribution of soluble fibrinogen vs insoluble fibrin matrices to vascular integrity, tissue repair, inflammation, and disease has been undefined and unapproachable. To establish the means to distinguish fibrinogen- and fibrin-dependent processes in vivo, Fib(AEK) mice were generated that carry normal levels of circulating fibrinogen but lack the capacity for fibrin polymer formation due to a germ-line mutation in the Aα chain thrombin cleavage site. Homozygous Fib(AEK) mice developed to term and exhibited postnatal survival superior to that of fibrinogen-deficient mice. Unlike fibrinogen-deficient mice, platelet-rich plasma from Fib(AEK) mice supported normal platelet aggregation in vitro, highlighting that fibrinogen(AEK) retains the functional capacity to support interactions with platelets. Thrombin failed to release fibrinopeptide-A from fibrinogen(AEK) and failed to induce polymer formation with Fib(AEK) plasma or purified fibrinogen(AEK) in 37°C mixtures regardless of incubation time. Fib(AEK) mice displayed both an absence of fibrin polymer formation following liver injury, as assessed by electron microscopy, and a failure to generate stable occlusive thrombi following FeCl3 injury of carotid arteries. Fib(AEK) mice exhibited a profound impediment in Staphylococcus aureus clearance following intraperitoneal infection similar to fibrinogen-deficient mice, yet Fib(AEK) mice displayed a significant infection dose-dependent survival advantage over fibrinogen-deficient mice following peritonitis challenge. Collectively, these findings establish for the first time that fibrin polymer is the molecular form critical for antimicrobial mechanisms while simultaneously highlighting biologically meaningful contributions and functions of the soluble molecule.


Asunto(s)
Fibrina/metabolismo , Fibrinógeno/fisiología , Interacciones Huésped-Patógeno , Mutación/genética , Peritonitis/etiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Pruebas de Coagulación Sanguínea , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Hemostáticos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Peritonitis/patología , Agregación Plaquetaria , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
9.
Ann Neurol ; 75(2): 303-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24740641

RESUMEN

Although multiple sclerosis (MS) has been associated with the coagulation system, the temporal and spatial regulation of coagulation activity in neuroinflammatory lesions is unknown. Using a novel molecular probe, we characterized the activity pattern of thrombin, the central protease of the coagulation cascade, in experimental autoimmune encephalomyelitis. Thrombin activity preceded onset of neurological signs, increased at disease peak, and correlated with fibrin deposition, microglial activation, demyelination, axonal damage, and clinical severity. Mice with a genetic deficit in prothrombin confirmed the specificity of the thrombin probe. Thrombin activity might be exploited for developing sensitive probes for preclinical detection and monitoring of neuroinflammation and MS progression.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Trombina/metabolismo , Animales , Axones/patología , Factores de Coagulación Sanguínea/química , Conexina 30 , Conexinas/genética , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Fibrina/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Poli I-C/toxicidad , Trombina/química
10.
Blood ; 121(10): 1783-94, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23299312

RESUMEN

Fibrinogen can support host antimicrobial containment/clearance mechanisms, yet selected pathogens appear to benefit from host procoagulants to drive bacterial virulence. Here, we explored the hypothesis that host fibrin(ogen), on balance, supports Staphylococcus aureus infection in the context of septicemia. Survival studies following intravenous infection in control and fibrinogen-deficient mice established the overall utility of host fibrin(ogen) to S. aureus virulence. Complementary studies in mice expressing mutant forms of fibrinogen-retaining clotting function, but lacking either the bacterial ClfA (Fibγ(Δ5)) binding motif or the host leukocyte integrin receptor αMß2 (Fibγ(390-396A)) binding motif, revealed the preeminent importance of the bacterial ClfA-fibrin(ogen) interaction in determining host survival. Studies of mice lacking platelets or the platelet integrin receptor subunit αIIb established that the survival benefits observed in Fibγ(Δ5) mice were largely independent of platelet αIIbß3-mediated engagement of fibrinogen. Fibγ(Δ5) mice exhibited reduced bacterial burdens in the hearts and kidneys, a blunted host proinflammatory cytokine response, diminished microscopic tissue damage, and significantly diminished plasma markers of cardiac and other organ damage. These findings indicate that host fibrin(ogen) and bacterial ClfA are dual determinants of virulence and that therapeutic interventions at the level of fibrinogen could be advantageous in S. aureus septicemia.


Asunto(s)
Coagulasa/metabolismo , Fibrinógeno/fisiología , Antígeno de Macrófago-1/fisiología , Sepsis/mortalidad , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Virulencia/fisiología , Afibrinogenemia/etiología , Animales , Adhesión Bacteriana/fisiología , Sitios de Unión , Citocinas/sangre , Citometría de Flujo , Ratones , Ratones Noqueados , Sepsis/etiología , Sepsis/prevención & control , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/metabolismo , Tasa de Supervivencia
11.
Hum Mol Genet ; 21(9): 1989-2004, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22381526

RESUMEN

In Duchenne muscular dystrophy (DMD), a persistently altered and reorganizing extracellular matrix (ECM) within inflamed muscle promotes damage and dysfunction. However, the molecular determinants of the ECM that mediate inflammatory changes and faulty tissue reorganization remain poorly defined. Here, we show that fibrin deposition is a conspicuous consequence of muscle-vascular damage in dystrophic muscles of DMD patients and mdx mice and that elimination of fibrin(ogen) attenuated dystrophy progression in mdx mice. These benefits appear to be tied to: (i) a decrease in leukocyte integrin α(M)ß(2)-mediated proinflammatory programs, thereby attenuating counterproductive inflammation and muscle degeneration; and (ii) a release of satellite cells from persistent inhibitory signals, thereby promoting regeneration. Remarkably, Fib-gamma(390-396A) (Fibγ(390-396A)) mice expressing a mutant form of fibrinogen with normal clotting function, but lacking the α(M)ß(2) binding motif, ameliorated dystrophic pathology. Delivery of a fibrinogen/α(M)ß(2) blocking peptide was similarly beneficial. Conversely, intramuscular fibrinogen delivery sufficed to induce inflammation and degeneration in fibrinogen-null mice. Thus, local fibrin(ogen) deposition drives dystrophic muscle inflammation and dysfunction, and disruption of fibrin(ogen)-α(M)ß(2) interactions may provide a novel strategy for DMD treatment.


Asunto(s)
Fibrina/metabolismo , Antígeno de Macrófago-1/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animales , Matriz Extracelular/metabolismo , Fibrinógeno/antagonistas & inhibidores , Fibrinógeno/genética , Fibrinógeno/metabolismo , Fibrinógeno/farmacología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Leucocitos/metabolismo , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Ratones Mutantes , Modelos Biológicos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/fisiología
12.
Blood ; 119(4): 1064-74, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22134166

RESUMEN

Circulating platelets contain high concentrations of TGF-ß1 in their α-granules and release it on platelet adhesion/activation. We hypothesized that uncontrolled in vitro release of platelet TGF-ß1 may confound measurement of plasma TGF-ß1 in mice and that in vivo release and activation may contribute to cardiac pathology in response to constriction of the transverse aorta, which produces both high shear and cardiac pressure overload. Plasma TGF-ß1 levels in blood collected from C57Bl/6 mice by the standard retro-bulbar technique were much higher than those obtained when prostaglandin E1 was added to inhibit release or when blood was collected percutaneously from the left ventricle under ultrasound guidance. Even with optimal blood drawing, plasma TGF-ß1 was lower in mice rendered profoundly thrombocytopenic or mice with selectively low levels of platelet TGF-ß1 because of megakaryocyte-specific disruption of their TGF-ß1 gene (Tgfb1(flox)). Tgfb1(flox) mice were also partially protected from developing cardiac hypertrophy, fibrosis, and systolic dysfunction in response to transverse aortic constriction. These studies demonstrate that plasma TGF-ß1 levels can be assessed accurately, but it requires special precautions; that platelet TGF-ß1 contributes to plasma levels of TGF-ß1; and that platelet TGF-ß1 contributes to the pathologic cardiac changes that occur in response to aortic constriction.


Asunto(s)
Plaquetas/metabolismo , Corazón/fisiopatología , Hipertensión/fisiopatología , Miocardio/patología , Factor de Crecimiento Transformador beta1/metabolismo , Alprostadil/metabolismo , Animales , Plaquetas/patología , Vasos Sanguíneos/patología , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/patología , Hemorragia/etiología , Hipertensión/sangre , Hipertensión/metabolismo , Hipertensión/patología , Integrasas/genética , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregación Plaquetaria , Anomalías Cutáneas/etiología , Trombocitopenia/etiología , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/genética
13.
J Exp Med ; 204(3): 571-82, 2007 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-17339406

RESUMEN

Perivascular microglia activation is a hallmark of inflammatory demyelination in multiple sclerosis (MS), but the mechanisms underlying microglia activation and specific strategies to attenuate their activation remain elusive. Here, we identify fibrinogen as a novel regulator of microglia activation and show that targeting of the interaction of fibrinogen with the microglia integrin receptor Mac-1 (alpha(M)beta(2), CD11b/CD18) is sufficient to suppress experimental autoimmune encephalomyelitis in mice that retain full coagulation function. We show that fibrinogen, which is deposited perivascularly in MS plaques, signals through Mac-1 and induces the differentiation of microglia to phagocytes via activation of Akt and Rho. Genetic disruption of fibrinogen-Mac-1 interaction in fibrinogen-gamma(390-396A) knock-in mice or pharmacologically impeding fibrinogen-Mac-1 interaction through intranasal delivery of a fibrinogen-derived inhibitory peptide (gamma(377-395)) attenuates microglia activation and suppresses relapsing paralysis. Because blocking fibrinogen-Mac-1 interactions affects the proinflammatory but not the procoagulant properties of fibrinogen, targeting the gamma(377-395) fibrinogen epitope could represent a potential therapeutic strategy for MS and other neuroinflammatory diseases associated with blood-brain barrier disruption and microglia activation.


Asunto(s)
Fibrina/fisiología , Fibrinógeno/fisiología , Microglía/inmunología , Microglía/patología , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Esclerosis Múltiple Recurrente-Remitente/prevención & control , Fragmentos de Péptidos/fisiología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Femenino , Antígeno de Macrófago-1/metabolismo , Antígeno de Macrófago-1/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Datos de Secuencia Molecular , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología
14.
Blood ; 118(10): 2889-95, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21788337

RESUMEN

Thrombomodulin (TM) is a predominantly endothelial transmembrane glycoprotein that modulates hemostatic function through a domain that controls thrombin-mediated proteolysis and an N-terminal lectin-like domain that controls inflammatory processes. To test the hypothesis that TM is a determinant of malignancy and dissect the importance of these functional domains in cancer biology, metastatic potential was evaluated in TM(Pro) mice expressing a mutant form of TM with reduced thrombin affinity and TM(LeD) mice lacking the N-terminal lectin-like domain. Studies of TM(Pro) mice revealed that TM is a powerful determinant of hematogenous metastasis. TM(Pro) mice exhibited a strongly prometastatic phenotype relative to control mice that was found to result from increased survival of tumor cells newly localized to the lung rather than any alteration in tumor growth. The impact of the TM(Pro) mutation on metastasis was dependent on both tumor cell-associated tissue factor and thrombin procoagulant function. In contrast, expression of a mutant form of TM lacking the lectin-like domain had no significant impact on metastasis. These studies directly demonstrate for the first time that TM-mediated regulation of tumor cell-driven procoagulant function strongly influences metastatic potential and suggest that endothelial cell-associated modulators of hemostasis may represent novel therapeutic targets in limiting tumor dissemination.


Asunto(s)
Lectinas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Mutación/genética , Trombina/metabolismo , Trombomodulina/fisiología , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Femenino , Hirudinas/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Neoplásicas Circulantes , Oligonucleótidos Antisentido/farmacología , Recuento de Plaquetas , Protrombina/antagonistas & inhibidores , Protrombina/genética , Proteínas Recombinantes/metabolismo , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patología
15.
Blood ; 117(23): 6326-37, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21436072

RESUMEN

Thrombin is a positive mediator of thrombus formation through the proteolytic activation of protease-activated receptors (PARs), fibrinogen, factor XI (fXI), and other substrates, and a negative regulator through activation of protein C, a natural anticoagulant with anti-inflammatory/cytoprotective properties. Protease-engineering studies have established that 2 active-site substitutions, W215A and E217A (fII(WE)), result in dramatically reduced catalytic efficiency with procoagulant substrates while largely preserving thrombomodulin (TM)-dependent protein C activation. To explore the hypothesis that a prothrombin variant favoring antithrombotic pathways would be compatible with development but limit inflammatory processes in vivo, we generated mice carrying the fII(WE) mutations within the endogenous prothrombin gene. Unlike fII-null embryos, fII(WE/WE) mice uniformly developed to term. Nevertheless, these mice ultimately succumbed to spontaneous bleeding events shortly after birth. Heterozygous fII(WT/WE) mice were viable and fertile despite a shift toward an antithrombotic phenotype exemplified by prolonged tail-bleeding times and times-to-occlusion after FeCl3 vessel injury. More interestingly, prothrombin(WE) expression significantly ameliorated the development of inflammatory joint disease in mice challenged with collagen-induced arthritis (CIA). The administration of active recombinant thrombin(WE) also suppressed the development of CIA in wild-type mice. These studies provide a proof-of-principle that pro/thrombin variants engineered with altered substrate specificity may offer therapeutic opportunities for limiting inflammatory disease processes.


Asunto(s)
Artritis Experimental/metabolismo , Mutación Missense , Protrombina/metabolismo , Sustitución de Aminoácidos , Animales , Artritis Experimental/genética , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Ratones , Ratones Mutantes , Proteína C/genética , Proteína C/metabolismo , Protrombina/genética
16.
J Biol Chem ; 286(11): 9865-74, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21247890

RESUMEN

In addition to its pivotal role in hemostasis, fibrinogen (Fg) and provisional fibrin matrices play important roles in inflammation and regulate innate immune responses by interacting with leukocytes. Efb (the extracellular fibrinogen-binding protein) is a secreted Staphylococcus aureus protein that engages host Fg and complement C3. However, the molecular details underlying the Efb-Fg interaction and the biological relevance of this interaction have not been determined. In the present study, we characterize the interaction of Efb with Fg. We demonstrate that the Fg binding activity is located within the intrinsically disordered N-terminal half of Efb (Efb-N) and that the D fragment of Fg is the region that mediates Efb-N binding. More detailed studies of the Efb-N-Fg interactions using ELISA and surface plasmon resonance analyses revealed that Efb-N exhibits a much higher affinity for Fg than typically observed with Fg-binding MSCRAMMs (microbial surface components recognizing adhesive matrix molecules), and data obtained from ELISA analyses using truncated Efb-N constructs demonstrate that Efb-N contains two binding sites located within residues 30-67 and 68-98, respectively. Efb-N inhibits neutrophil adhesion to immobilized Fg by binding to Fg and blocking the interaction of the protein with the leukocyte integrin receptor, α(M)ß(2). A motif in the Fg γ chain previously shown to be central to the α(M)ß(2) interaction was shown to be functionally distinguishable from the Efb-N binding site, suggesting that the Fg-Efb interaction indirectly impedes Fg engagement by α(M)ß(2). Taken together, these studies provide insights into how Efb interacts with Fg and suggest that Efb may support bacterial virulence at least in part by impeding Fg-driven leukocyte adhesion events.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fibrinógeno/metabolismo , Neutrófilos/metabolismo , Staphylococcus aureus/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/química , Sitios de Unión , Adhesión Celular , Fibrinógeno/química , Células HEK293 , Humanos , Neutrófilos/química , Unión Proteica , Staphylococcus aureus/química , Staphylococcus aureus/patogenicidad
17.
Blood ; 116(24): 5191-9, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20876456

RESUMEN

It has been recognized for nearly 80 years that insoluble aluminum salts are good immunologic adjuvants and that they form long-lived nodules in vivo. Nodule formation has long been presumed to be central for adjuvant activity by providing an antigen depot, but the composition and function of these nodules is poorly understood. We show here that aluminum salt nodules formed within hours of injection and contained the clotting protein fibrinogen. Fibrinogen was critical for nodule formation and required processing to insoluble fibrin by thrombin. DNase treatment partially disrupted the nodules, and the nodules contained histone H3 and citrullinated H3, features consistent with extracellular traps. Although neutrophils were not essential for nodule formation, CD11b(+) cells were implicated. Vaccination of fibrinogen-deficient mice resulted in normal CD4 T-cell and antibody responses and enhanced CD8 T-cell responses, indicating that nodules are not required for aluminum's adjuvant effect. Moreover, the ability of aluminum salts to retain antigen in the body, the well-known depot effect, was unaffected by the absence of nodules. We conclude that aluminum adjuvants form fibrin-dependent nodules in vivo, that these nodules have properties of extracellular traps, and the nodules are not required for aluminum salts to act as adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Aluminio/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Aluminio/farmacología , Animales , Fibrina/metabolismo , Fibrinógeno/metabolismo , Histonas/metabolismo , Ratones , Ratones Noqueados , Sales (Química) , Linfocitos T/inmunología , Trombina/metabolismo , Vacunación
18.
J Neurosci ; 30(17): 5843-54, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20427645

RESUMEN

Scar formation in the nervous system begins within hours after traumatic injury and is characterized primarily by reactive astrocytes depositing proteoglycans that inhibit regeneration. A fundamental question in CNS repair has been the identity of the initial molecular mediator that triggers glial scar formation. Here we show that the blood protein fibrinogen, which leaks into the CNS immediately after blood-brain barrier (BBB) disruption or vascular damage, serves as an early signal for the induction of glial scar formation via the TGF-beta/Smad signaling pathway. Our studies revealed that fibrinogen is a carrier of latent TGF-beta and induces phosphorylation of Smad2 in astrocytes that leads to inhibition of neurite outgrowth. Consistent with these findings, genetic or pharmacologic depletion of fibrinogen in mice reduces active TGF-beta, Smad2 phosphorylation, glial cell activation, and neurocan deposition after cortical injury. Furthermore, stereotactic injection of fibrinogen into the mouse cortex is sufficient to induce astrogliosis. Inhibition of the TGF-beta receptor pathway abolishes the fibrinogen-induced effects on glial scar formation in vivo and in vitro. These results identify fibrinogen as a primary astrocyte activation signal, provide evidence that deposition of inhibitory proteoglycans is induced by a blood protein that leaks in the CNS after vasculature rupture, and point to TGF-beta as a molecular link between vascular permeability and scar formation.


Asunto(s)
Astrocitos/fisiología , Corteza Cerebral/lesiones , Corteza Cerebral/fisiopatología , Cicatriz/fisiopatología , Fibrinógeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/irrigación sanguínea , Fibrinógeno/genética , Gliosis/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuritas/fisiología , Neurocano , Fosforilación , Proteoglicanos/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo
19.
Kidney Int ; 80(10): 1035-44, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21734641

RESUMEN

Fibrinogen plays an important role in blood coagulation but its function extends far beyond blood clotting being involved in inflammation and repair. Besides these crucial functions it can also promote tissue fibrosis. To determine whether fibrinogen is involved in the development of renal tubulointerstitial fibrosis we utilized the profibrotic model of unilateral ureteral obstruction in fibrinogen-deficient mice. In the heterozygotes, obstruction was associated with a massive deposition of intrarenal fibrinogen. Fibrinogen deficiency provided significant protection from interstitial damage and tubular disruption, attenuated collagen accumulation, and greatly reduced de novo expression of α-smooth muscle actin in the obstructed kidney. While no differences were found in renal inflammatory cell infiltration, fibrinogen deficiency was associated with a significant reduction in interstitial cell proliferation, a hallmark of renal fibrosis. In vitro, fibrinogen directly stimulated renal fibroblast proliferation in a dose-dependent manner. This mitogenic effect of fibrinogen was mediated by at least three different cell surface receptors on renal fibroblasts: TLR2, TLR4, and ICAM-1. Thus, our study suggests that fibrinogen promotes renal fibrosis by triggering resident fibroblast proliferation.


Asunto(s)
Proliferación Celular , Fibrinógeno/metabolismo , Fibroblastos/metabolismo , Túbulos Renales/metabolismo , Obstrucción Ureteral/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrinógeno/genética , Fibroblastos/patología , Fibrosis , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Túbulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Interferencia de ARN , Ratas , Factores de Tiempo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Transfección , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
20.
Blood ; 113(6): 1358-64, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19056689

RESUMEN

Bacterial plasminogen activators are commonplace among microbial pathogens, implying a central role of host plasmin in supporting bacterial virulence. Group A streptococci (GAS) secrete streptokinase, a specific activator of human plasminogen (PLG). The critical contribution of the streptokinase-PLG interaction to GAS pathogenicity was recently demonstrated using mice expressing human PLG. To examine the importance of thrombin generation in antimicrobial host defense, we challenged mice with deficiency of factor V (FV) in either the plasma or platelet compartment. Reduction of FV in either pool resulted in markedly increased mortality after GAS infection, with comparison to heterozygous F5-deficient mice suggesting a previously unappreciated role for the platelet FV pool in host defense. Mice with complete deficiency of fibrinogen also demonstrated markedly increased mortality to GAS infection relative to controls. Although FV Leiden may be protective in the setting of severe sepsis in humans, no significant survival advantage was observed in GAS-infected mice carrying the FV Leiden mutation. Taken together, our data support the hypothesis that local thrombosis/fibrin deposition limits the survival and dissemination of at least a subset of microbial pathogens and suggest that common variation in hemostatic factors among humans could affect host susceptibility to a variety of infectious diseases.


Asunto(s)
Susceptibilidad a Enfermedades , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Trombina/metabolismo , Animales , Factor V/fisiología , Femenino , Fibrinógeno/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Tiempo de Protrombina , Streptococcus pyogenes/crecimiento & desarrollo , Estreptoquinasa/metabolismo , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA