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










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(1): e1010227, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35041705

RESUMO

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.


Assuntos
Fibrinogênio/imunologia , Peritonite/imunologia , Infecções Estafilocócicas/imunologia , Animais , Coagulase/imunologia , Coagulase/metabolismo , Fibrina/metabolismo , Camundongos , Peritonite/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33443167

RESUMO

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.


Assuntos
Fibrinogênio/metabolismo , Peritonite/metabolismo , Protrombina/metabolismo , Animais , Antibacterianos/metabolismo , Anti-Infecciosos/metabolismo , Coagulação Sanguínea , Coagulase/metabolismo , Feminino , Fibrina/metabolismo , Fibrinogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Tromboplastina
3.
PLoS Genet ; 15(3): e1007948, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30870413

RESUMO

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.


Assuntos
Antitrombina III/genética , Cofator II da Heparina/genética , Junção Neuromuscular/genética , Protrombina/genética , Sinapses/genética , Animais , Perfilação da Expressão Gênica , Camundongos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Músculo Esquelético/metabolismo , Degeneração Neural/genética , Neuroglia , Junção Neuromuscular/crescimento & desenvolvimento , Células de Schwann/metabolismo , Trombina/genética
4.
J Clin Invest ; 128(8): 3356-3368, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29723163

RESUMO

Hemostasis requires conversion of fibrinogen to fibrin fibers that generate a characteristic network, interact with blood cells, and initiate tissue repair. The fibrin network is porous and highly permeable, but the spatial arrangement of the external clot face is unknown. Here we show that fibrin transitioned to the blood-air interface through Langmuir film formation, producing a protective film confining clots in human and mouse models. We demonstrated that only fibrin is required for formation of the film, and that it occurred in vitro and in vivo. The fibrin film connected to the underlying clot network through tethering fibers. It was digested by plasmin, and formation of the film was prevented with surfactants. Functionally, the film retained blood cells and protected against penetration by bacterial pathogens in a murine model of dermal infection. Our data show a remarkable aspect of blood clotting in which fibrin forms a protective film covering the external surface of the clot, defending the organism against microbial invasion.


Assuntos
Bactérias/genética , Fenômenos Fisiológicos Bacterianos , Biofilmes , Coagulação Sanguínea , Fibrina/metabolismo , Dermatopatias Bacterianas/metabolismo , Animais , Bactérias/patogenicidade , Modelos Animais de Doenças , Humanos , Camundongos , Dermatopatias Bacterianas/microbiologia
5.
J Neurosci ; 37(14): 3776-3788, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28275164

RESUMO

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.


Assuntos
Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/prevenção & controle , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Paralisia/metabolismo , Paralisia/prevenção & controle , Plasminogênio/deficiência , Animais , Células Cultivadas , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Paralisia/patologia
6.
J Bone Miner Res ; 32(2): 294-308, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27530373

RESUMO

Extensive or persistent calcium phosphate deposition within soft tissues after severe traumatic injury or major orthopedic surgery can result in pain and loss of joint function. The pathophysiology of soft tissue calcification, including dystrophic calcification and heterotopic ossification (HO), is poorly understood; consequently, current treatments are suboptimal. Here, we show that plasmin protease activity prevents dystrophic calcification within injured skeletal muscle independent of its canonical fibrinolytic function. After muscle injury, dystrophic calcifications either can be resorbed during the process of tissue healing, persist, or become organized into mature bone (HO). Without sufficient plasmin activity, dystrophic calcifications persist after muscle injury and are sufficient to induce HO. Downregulating the primary inhibitor of plasmin (α2-antiplasmin) or treating with pyrophosphate analogues prevents dystrophic calcification and subsequent HO in vivo. Because plasmin also supports bone homeostasis and fracture repair, increasing plasmin activity represents the first pharmacologic strategy to prevent soft tissue calcification without adversely affecting systemic bone physiology or concurrent muscle and bone regeneration. © 2016 American Society for Bone and Mineral Research.


Assuntos
Calcinose/metabolismo , Fibrinolisina/metabolismo , Músculo Esquelético/lesões , Animais , Calcinose/tratamento farmacológico , Calcinose/genética , Cardiotoxinas , Difosfatos/farmacologia , Difosfatos/uso terapêutico , Fibrinolisina/deficiência , Fibrinólise/efeitos dos fármacos , Predisposição Genética para Doença , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/patologia , Regeneração/efeitos dos fármacos
7.
Blood ; 128(5): 721-31, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27252233

RESUMO

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.


Assuntos
Precursores Enzimáticos/metabolismo , Hemostasia , Protrombina/metabolismo , Trombina/metabolismo , Alelos , Animais , Coagulação Sanguínea , Retração do Coágulo , Venenos de Crotalídeos , Embrião de Mamíferos/metabolismo , Fibrose , Metaloendopeptidases , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Metástase Neoplásica , Agregação Plaquetária , Análise de Sobrevida , Trombose/metabolismo , Trombose/patologia
8.
Nat Commun ; 6: 8164, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26353940

RESUMO

Autoimmunity and macrophage recruitment into the central nervous system (CNS) are critical determinants of neuroinflammatory diseases. However, the mechanisms that drive immunological responses targeted to the CNS remain largely unknown. Here we show that fibrinogen, a central blood coagulation protein deposited in the CNS after blood-brain barrier disruption, induces encephalitogenic adaptive immune responses and peripheral macrophage recruitment into the CNS leading to demyelination. Fibrinogen stimulates a unique transcriptional signature in CD11b(+) antigen-presenting cells inducing the recruitment and local CNS activation of myelin antigen-specific Th1 cells. Fibrinogen depletion reduces Th1 cells in the multiple sclerosis model, experimental autoimmune encephalomyelitis. Major histocompatibility complex (MHC) II-dependent antigen presentation, CXCL10- and CCL2-mediated recruitment of T cells and macrophages, respectively, are required for fibrinogen-induced encephalomyelitis. Inhibition of the fibrinogen receptor CD11b/CD18 protects from all immune and neuropathologic effects. Our results show that the final product of the coagulation cascade is a key determinant of CNS autoimmunity.


Assuntos
Autoimunidade/imunologia , Encéfalo/imunologia , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Fibrinogênio/imunologia , Genes MHC da Classe II/imunologia , Macrófagos/imunologia , Medula Espinal/imunologia , Células Th1/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Imunidade Adaptativa/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Autoimunidade/efeitos dos fármacos , Autoimunidade/genética , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Receptor 1 de Quimiocina CX3C , Proliferação de Células , Quimiocina CCL2/imunologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quimiocinas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Doenças Desmielinizantes/genética , Fibrina , Fibrinogênio/farmacologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Genes MHC da Classe II/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microglia , Glicoproteína Mielina-Oligodendrócito/imunologia , Ratos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
10.
Blood ; 126(15): 1844-55, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26286849

RESUMO

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.


Assuntos
Anemia Falciforme/complicações , Terapia Genética , Hipertensão Pulmonar/prevenção & controle , Inflamação/prevenção & controle , Protrombina/fisiologia , Doenças Vasculares/prevenção & controle , Anemia Falciforme/mortalidade , Anemia Falciforme/fisiopatologia , Animais , Coagulação Sanguínea , Células Cultivadas , Hipertensão Pulmonar/etiologia , Técnicas Imunoenzimáticas , Inflamação/etiologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Oligorribonucleotídeos Antissenso/farmacologia , Protrombina/antagonistas & inibidores , Taxa de Sobrevida , Trombina/metabolismo , Doenças Vasculares/etiologia
11.
Blood ; 126(17): 2047-58, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26228483

RESUMO

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.


Assuntos
Fibrina/metabolismo , Fibrinogênio/fisiologia , Interações Hospedeiro-Patógeno , Mutação/genética , Peritonite/etiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Testes de Coagulação Sanguínea , Células Cultivadas , Citometria de Fluxo , Perfilação da Expressão Gênica , Hemostáticos , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Peritonite/patologia , Agregação Plaquetária , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
12.
J Clin Invest ; 125(8): 3117-31, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26214526

RESUMO

Bone formation during fracture repair inevitably initiates within or around extravascular deposits of a fibrin-rich matrix. In addition to a central role in hemostasis, fibrin is thought to enhance bone repair by supporting inflammatory and mesenchymal progenitor egress into the zone of injury. However, given that a failure of efficient fibrin clearance can impede normal wound repair, the precise contribution of fibrin to bone fracture repair, whether supportive or detrimental, is unknown. Here, we employed mice with genetically and pharmacologically imposed deficits in the fibrin precursor fibrinogen and fibrin-degrading plasminogen to explore the hypothesis that fibrin is vital to the initiation of fracture repair, but impaired fibrin clearance results in derangements in bone fracture repair. In contrast to our hypothesis, fibrin was entirely dispensable for long-bone fracture repair, as healing fractures in fibrinogen-deficient mice were indistinguishable from those in control animals. However, failure to clear fibrin from the fracture site in plasminogen-deficient mice severely impaired fracture vascularization, precluded bone union, and resulted in robust heterotopic ossification. Pharmacological fibrinogen depletion in plasminogen-deficient animals restored a normal pattern of fracture repair and substantially limited heterotopic ossification. Fibrin is therefore not essential for fracture repair, but inefficient fibrinolysis decreases endochondral angiogenesis and ossification, thereby inhibiting fracture repair.


Assuntos
Fibrinólise , Consolidação da Fratura , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/prevenção & controle , Animais , Fibrina/genética , Fibrina/metabolismo , Fibrinogênio/genética , Fibrinogênio/metabolismo , Camundongos , Camundongos Knockout , Ossificação Heterotópica/genética , Plasminogênio/genética , Plasminogênio/metabolismo
13.
Blood ; 125(3): 427-37, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25336631

RESUMO

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.


Assuntos
Artrite Experimental/patologia , Doenças Ósseas/fisiopatologia , Fator XIII/fisiologia , Inflamação/fisiopatologia , Osteoclastos/patologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/imunologia , Western Blotting , Doenças Ósseas/complicações , Diferenciação Celular , Células Cultivadas , Colágeno/toxicidade , Feminino , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Osteoclastos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Clin Invest ; 124(8): 3590-600, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24983320

RESUMO

Venous thrombi, fibrin- and rbc-rich clots triggered by inflammation and blood stasis, underlie devastating, and sometimes fatal, occlusive events. During intravascular fibrin deposition, rbc are thought to become passively trapped in thrombi and therefore have not been considered a modifiable thrombus component. In the present study, we determined that activity of the transglutaminase factor XIII (FXIII) is critical for rbc retention within clots and directly affects thrombus size. Compared with WT mice, mice carrying a homozygous mutation in the fibrinogen γ chain (Fibγ390-396A) had a striking 50% reduction in thrombus weight due to reduced rbc content. Fibrinogen from mice harboring the Fibγ390-396A mutation exhibited reduced binding to FXIII, and plasma from these mice exhibited delayed FXIII activation and fibrin crosslinking, indicating these residues mediate FXIII binding and activation. FXIII-deficient mice phenocopied mice carrying Fibγ390-396A and produced smaller thrombi with fewer rbc than WT mice. Importantly, FXIII-deficient human clots also exhibited reduced rbc retention. The addition of FXIII to FXIII-deficient clots increased rbc retention, while inhibition of FXIII activity in normal blood reduced rbc retention and produced smaller clots. These findings establish the FXIII-fibrinogen axis as a central determinant in venous thrombogenesis and identify FXIII as a potential therapeutic target for limiting venous thrombosis.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/patologia , Fator XIII/metabolismo , Fibrinogênio/genética , Fibrinogênio/metabolismo , Trombose Venosa/sangue , Animais , Coagulação Sanguínea , Modelos Animais de Doenças , Deficiência do Fator XIII/sangue , Deficiência do Fator XIII/genética , Fator XIIIa/genética , Fator XIIIa/metabolismo , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Proteínas Mutantes/genética , Mutação , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Trombose Venosa/genética
15.
Ann Neurol ; 75(2): 303-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24740641

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Trombina/metabolismo , Animais , Axônios/patologia , Fatores de Coagulação Sanguínea/química , Conexina 30 , Conexinas/genética , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/induzido quimicamente , Fibrina/metabolismo , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Básica da Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/toxicidade , Poli I-C/toxicidade , Trombina/química
16.
Arthritis Rheumatol ; 66(8): 2222-33, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24664548

RESUMO

OBJECTIVE: Osteoporosis is a skeletal disorder characterized by low bone mass and increased bone fragility associated with aging, menopause, smoking, obesity, or diabetes. Persistent inflammation has been identified as an instigating factor in progressive bone loss. In addition to the role of fibrin in coagulation, inordinate fibrin deposition within a tissue matrix results in increased local inflammation. Given that fibrin accumulation is a hallmark of osteoporosis-related comorbidities, we undertook this study to test the hypothesis that persistent fibrin deposition causes inflammatory osteoporosis. METHODS: Multiple imaging modalities, bone integrity metrics, and histologic analyses were employed to evaluate skeletal derangements in relation to fibrin deposition, circulating fibrinogen levels, and systemic markers of inflammation in mice that were plasminogen deficient and in plasminogen-deficient mice that were concomitantly either fibrinogen deficient or carrying a mutant form of fibrinogen lacking the αM ß2 binding motif. RESULTS: Mice generated with a genetic deficit in the key fibrinolytic protease, plasmin, uniformly developed severe osteoporosis. Furthermore, the development of osteoporosis was fibrin(ogen) dependent, and the derangements in the bone remodeling unit were mechanistically tied to fibrin(ogen)-mediated activation of osteoclasts via activation of the leukocyte integrin receptor αM ß2 on monocytes and secondary stimulation of osteoblasts by RANKL. Notably, the genetic elimination of fibrin(ogen) or the expression of a mutant form of fibrinogen retaining clotting function but lacking the αM ß2 binding motif prevented the degenerative skeletal phenotypes, resulting in normal local and systemic cytokine levels. CONCLUSION: Taken together, these data reveal for the first time that fibrin promotes inflammation-driven systemic osteoporosis, which suggests a novel association between hemostasis, inflammation, and bone biology.


Assuntos
Fibrina/metabolismo , Fibrinolisina/fisiologia , Fibrinólise/fisiologia , Inflamação/etiologia , Osteoporose/etiologia , Animais , Masculino , Camundongos
17.
Arthritis Rheumatol ; 66(6): 1504-16, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24574269

RESUMO

OBJECTIVE: A fundamental metric in the diagnosis of arthropathies is the pattern of joint involvement, including differences in proximal versus distal joints and patterns of symmetric or asymmetric disease. The basis for joint selectivity among arthritides and/or within a defined disease such as rheumatoid arthritis remains enigmatic. Coagulation and fibrinolytic activity are observed in both experimental animals with inflammatory joint disease and patients with inflammatory arthritis. However, the contribution of specific hemostatic factors to joint disease is not fully defined. We sought to determine the contribution of the fibrinolytic protease, plasminogen, to tumor necrosis factor α (TNFα)-driven arthritis in distinct joints in mice. METHODS: The impact of plasminogen and/or fibrinogen genetic deficiencies on arthritis progression was evaluated in Tg197 mice genetically predisposed to spontaneous, nonabating, and erosive polyarthritis due to exuberant human TNFα expression. RESULTS: Elimination of plasminogen in Tg197 mice significantly exacerbated the incidence and severity of arthritis within the paw joints, but simultaneously and dramatically diminished the entire spectrum of pathologies within the knee joints of the same animals. These opposing outcomes were both mechanistically linked to fibrin(ogen), in that superimposing fibrinogen deficiency reversed both the proarthritic phenotype in the paws and arthritis resistance in the knees of plasminogen-deficient mice. Intriguingly, the change in disease severity in the knees, but not the paws, was associated with a plasminogen-dependent reduction in matrix metalloproteinase 9 activity. CONCLUSION: Plasminogen is a key molecular determinant of inflammatory joint disease capable of simultaneously driving or ameliorating arthritis pathogenesis in distinct anatomic locations in the same subject.


Assuntos
Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Articulações/metabolismo , Plasminogênio/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Fibrinogênio/genética , Fibrinogênio/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasminogênio/deficiência , Plasminogênio/genética , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/metabolismo
18.
PLoS One ; 8(2): e55784, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23409043

RESUMO

BACKGROUND: Variations in the blood coagulation activity, determined genetically or by medication, may alter atherosclerotic plaque progression, by influencing pleiotropic effects of coagulation proteases. Published experimental studies have yielded contradictory findings on the role of hypercoagulability in atherogenesis. We therefore sought to address this matter by extensively investigating the in vivo significance of genetic alterations and pharmacologic inhibition of thrombin formation for the onset and progression of atherosclerosis, and plaque phenotype determination. METHODOLOGY/PRINCIPAL FINDINGS: We generated transgenic atherosclerosis-prone mice with diminished coagulant or hypercoagulable phenotype and employed two distinct models of atherosclerosis. Gene-targeted 50% reduction in prothrombin (FII(-/WT):ApoE(-/-)) was remarkably effective in limiting disease compared to control ApoE(-/-) mice, associated with significant qualitative benefits, including diminished leukocyte infiltration, altered collagen and vascular smooth muscle cell content. Genetically-imposed hypercoagulability in TM(Pro/Pro):ApoE(-/-) mice resulted in severe atherosclerosis, plaque vulnerability and spontaneous atherothrombosis. Hypercoagulability was associated with a pronounced neutrophilia, neutrophil hyper-reactivity, markedly increased oxidative stress, neutrophil intraplaque infiltration and apoptosis. Administration of either the synthetic specific thrombin inhibitor Dabigatran etexilate, or recombinant activated protein C (APC), counteracted the pro-inflammatory and pro-atherogenic phenotype of pro-thrombotic TM(Pro/Pro):ApoE(-/-) mice. CONCLUSIONS/SIGNIFICANCE: We provide new evidence highlighting the importance of neutrophils in the coagulation-inflammation interplay during atherogenesis. Our findings reveal that thrombin-mediated proteolysis is an unexpectedly powerful determinant of atherosclerosis in multiple distinct settings. These studies suggest that selective anticoagulants employed to prevent thrombotic events may also be remarkably effective in clinically impeding the onset and progression of cardiovascular disease.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/genética , Aterosclerose/metabolismo , Neutrófilos/metabolismo , Trombina/metabolismo , Trombose/etiologia , Animais , Apolipoproteínas E/genética , Aterosclerose/complicações , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Dabigatrana , Modelos Animais de Doenças , Progressão da Doença , Feminino , Hematopoese , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , Fenótipo , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/patologia , Piridinas/administração & dosagem , Piridinas/farmacologia , Espécies Reativas de Oxigênio , Trombina/genética , Trombose/tratamento farmacológico
19.
Blood ; 121(10): 1783-94, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23299312

RESUMO

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.


Assuntos
Coagulase/metabolismo , Fibrinogênio/fisiologia , Antígeno de Macrófago 1/fisiologia , Sepse/mortalidade , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Virulência/fisiologia , Afibrinogenemia/etiologia , Animais , Aderência Bacteriana/fisiologia , Sítios de Ligação , Citocinas/sangue , Citometria de Fluxo , Camundongos , Camundongos Knockout , Sepse/etiologia , Sepse/prevenção & controle , Infecções Estafilocócicas/complicações , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/metabolismo , Taxa de Sobrevida
20.
Nat Commun ; 3: 1227, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23187627

RESUMO

Blood-brain barrier disruption, microglial activation and neurodegeneration are hallmarks of multiple sclerosis. However, the initial triggers that activate innate immune responses and their role in axonal damage remain unknown. Here we show that the blood protein fibrinogen induces rapid microglial responses toward the vasculature and is required for axonal damage in neuroinflammation. Using in vivo two-photon microscopy, we demonstrate that microglia form perivascular clusters before myelin loss or paralysis onset and that, of the plasma proteins, fibrinogen specifically induces rapid and sustained microglial responses in vivo. Fibrinogen leakage correlates with areas of axonal damage and induces reactive oxygen species release in microglia. Blocking fibrin formation with anticoagulant treatment or genetically eliminating the fibrinogen binding motif recognized by the microglial integrin receptor CD11b/CD18 inhibits perivascular microglial clustering and axonal damage. Thus, early and progressive perivascular microglial clustering triggered by fibrinogen leakage upon blood-brain barrier disruption contributes to axonal damage in neuroinflammatory disease.


Assuntos
Axônios/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Fibrinogênio/fisiologia , Microglia/patologia , Animais , Axônios/fisiologia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Fibrina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Microscopia de Fluorescência por Excitação Multifotônica , Espécies Reativas de Oxigênio/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...