Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Am J Physiol Cell Physiol ; 326(1): C40-C49, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37955120

RESUMEN

The blood-brain barrier is composed of microvascular endothelial cells, immune cells, and astrocytes that work in concert with the coagulation cascade to control inflammation and immune cell infiltration into the central nervous system. Endothelial cell dysfunction leading to increased permeability and compromised barrier function are hallmarks of neuroinflammatory and autoimmune disorders, including multiple sclerosis (MS). Therapeutic strategies that improve or protect endothelial barrier function may be beneficial in the treatment or prevention of neuroinflammatory diseases. We therefore tested the hypothesis that biasing thrombin toward anticoagulant and cytoprotective activities would provide equivalent or even additive benefit compared with standard-of-care therapeutic strategies, including corticosteroids. In a mouse model of relapsing-remitting MS, treatment with the thrombin mutant, E-WE thrombin, an engineered thrombin mutant with cytoprotective activities that is biased toward anticoagulant and cytoprotective activity, reduced neuroinflammation and extracellular fibrin formation in SJL mice inoculated with proteolipid protein (PLP) peptide. When administered at the onset of detectable disease, E-WE thrombin significantly improved the disease severity of the initial attack as well as the relapse and delayed the onset of relapse to a similar extent as observed with methylprednisolone. Both methylprednisolone and E-WE thrombin reduced demyelination and immune cell recruitment. These results provide rationale for considering engineered forms of thrombin biased toward anticoagulant and cytoprotective activity as a therapeutic strategy and perhaps an effective alternative to high-dose methylprednisolone for the management of acute relapsing MS attacks.NEW & NOTEWORTHY There are limited treatment options for mitigating acute relapsing attacks for patients with multiple sclerosis. We tested the hypothesis that harnessing the cytoprotective activity of the blood coagulation enzyme, thrombin, would provide benefit and protection against relapsing disease in a mouse model of MS. Our results provide rationale for considering engineered forms of thrombin biased toward cytoprotective activity as a therapeutic strategy and perhaps an alternative to steroids for the management of relapsing MS attacks.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Trombina , Animales , Humanos , Ratones , Anticoagulantes , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Metilprednisolona , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Gravedad del Paciente , Recurrencia , Trombina/uso terapéutico
2.
Blood ; 138(22): 2173-2184, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34086880

RESUMEN

End-stage renal disease (ESRD) patients on chronic hemodialysis have repeated blood exposure to artificial surfaces that can trigger clot formation within the hemodialysis circuit. Dialyzer clotting can lead to anemia despite erythropoietin and iron supplementation. Unfractionated heparin prevents clotting during hemodialysis, but it is not tolerated by all patients. Although heparin-free dialysis is performed, intradialytic blood entrapment can be problematic. To address this issue, we performed a randomized, double-blind, phase 2 study comparing AB023, a unique antibody that binds factor XI (FXI) and blocks its activation by activated FXII, but not by thrombin, to placebo in 24 patients with ESRD undergoing heparin-free hemodialysis. Patients were randomized to receive a single predialysis dose of AB023 (0.25 or 0.5 mg/kg) or placebo in a 2:1 ratio, and safety and preliminary efficacy were compared with placebo and observations made prior to dosing within each treatment arm. AB023 administration was not associated with impaired hemostasis or other drug-related adverse events. Occlusive events requiring hemodialysis circuit exchange were less frequent and levels of thrombin-antithrombin complexes and C-reactive protein were lower after AB023 administration compared with data collected prior to dosing. AB023 also reduced potassium and iron entrapment in the dialyzers, consistent with less blood accumulation within the dialyzers. We conclude that despite the small sample size, inhibition of contact activation-induced coagulation with AB023 was well tolerated and reduced clotting within the dialyzer. This trial was registered at www.clinicaltrials.gov as #NCT03612856.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antitrombinas/uso terapéutico , Fallo Renal Crónico/terapia , Diálisis Renal/métodos , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Antitrombinas/efectos adversos , Método Doble Ciego , Factor XI/antagonistas & inhibidores , Femenino , Hemostasis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Efecto Placebo , Diálisis Renal/efectos adversos , Trombosis/etiología , Trombosis/prevención & control
3.
Metab Brain Dis ; 38(7): 2383-2391, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37341855

RESUMEN

Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Masculino , Ratones , Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Factor XI/antagonistas & inhibidores , Factor XI/metabolismo , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Médula Espinal/metabolismo
4.
Blood ; 135(9): 689-699, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31977000

RESUMEN

Although thrombin is a key enzyme in the coagulation cascade and is required for both normal hemostasis and pathologic thrombogenesis, it also participates in its own negative feedback via activation of protein C, which downregulates thrombin generation by enzymatically inactivating factors Va and VIIIa. Our group and others have previously shown that thrombin's procoagulant and anticoagulant activities can be effectively disassociated to varying extents through site-directed mutagenesis. The thrombin mutant W215A/E217A (WE thrombin) has been one of the best characterized constructs with selective activity toward protein C. Although animal studies have demonstrated that WE thrombin acts as an anticoagulant through activated protein C (APC) generation, the observed limited systemic anticoagulation does not fully explain the antithrombotic potency of this or other thrombin mutants. AB002 (E-WE thrombin) is an investigational protein C activator thrombin analog in phase 2 clinical development (clinicaltrials.gov NCT03963895). Here, we demonstrate that this molecule is a potent enzyme that is able to rapidly interrupt arterial-type thrombus propagation at exceedingly low doses (<2 µg/kg, IV), yet without substantial systemic anticoagulation in baboons. We demonstrate that AB002 produces APC on platelet aggregates and competitively inhibits thrombin-activatable fibrinolysis inhibitor (carboxypeptidase B2) activation in vitro, which may contribute to the observed in vivo efficacy. We also describe its safety and activity in a phase 1 first-in-human clinical trial. Together, these results support further clinical evaluation of AB002 as a potentially safe and effective new approach for treating or preventing acute thrombotic and thromboembolic conditions. This trial was registered at www.clinicaltrials.gov as #NCT03453060.


Asunto(s)
Fibrinolíticos/farmacología , Proteína C/efectos de los fármacos , Trombina/análogos & derivados , Trombosis/prevención & control , Adulto , Animales , Método Doble Ciego , Humanos , Persona de Mediana Edad , Papio , Proteínas Recombinantes/farmacología
5.
Metab Brain Dis ; 36(3): 407-420, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33411219

RESUMEN

The coagulation cascade and immune system are intricately linked, highly regulated and respond cooperatively in response to injury and infection. Increasingly, evidence of hyper-coagulation has been associated with autoimmune disorders, including multiple sclerosis (MS). The pathophysiology of MS includes immune cell activation and recruitment to the central nervous system (CNS) where they degrade myelin sheaths, leaving neuronal axons exposed to damaging inflammatory mediators. Breakdown of the blood-brain barrier (BBB) facilitates the entry of peripheral immune cells. Evidence of thrombin activity has been identified within the CNS of MS patients and studies using animal models of experimental autoimmune encephalomyelitis (EAE), suggest increased thrombin generation and activity may play a role in the pathogenesis of MS as well as inhibit remyelination processes. Thrombin is a serine protease capable of cleaving multiple substrates, including protease activated receptors (PARs), fibrinogen, and protein C. Cleavage of all three of these substrates represent pathways through which thrombin activity may exert immuno-regulatory effects and regulate permeability of the BBB during MS and EAE. In this review, we summarize evidence that thrombin activity directly, through PARs, and indirectly, through fibrin formation and activation of protein C influences neuro-immune responses associated with MS and EAE pathology.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Trombina/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Humanos
6.
Arterioscler Thromb Vasc Biol ; 39(4): 799-809, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30700130

RESUMEN

Objective- Factor XI (FXI) contributes to thrombotic disease while playing a limited role in normal hemostasis. We generated a unique, humanized anti-FXI antibody, AB023, which blocks factor XIIa-mediated FXI activation without inhibiting FXI activation by thrombin or the procoagulant function of FXIa. We sought to confirm the antithrombotic activity of AB023 in a baboon thrombosis model and to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics in healthy adult subjects. Approach and Results- In a primate model of acute vascular graft thrombosis, AB023 reduced platelet and fibrin accumulation within the grafts by >75%. To evaluate the safety of AB023, we performed a first-in-human study in healthy adult volunteers without any serious adverse events. Overall, 10 of 21 (48%) subjects experienced 20 treatment-emergent adverse events, with 7 of 16 (44%) subjects following active treatment and 3 of 5 (60%) subjects following placebo. AB023 did not increase bleeding or prothrombin times. Anticoagulation was verified by a saturable ≈2-fold prolongation of the partial thromboplastin time for over 1 month after the highest dose. Conclusions- AB023, which inhibits contact activation-initiated blood coagulation in vitro and experimental thrombus formation in primates, produced a dose-dependent duration of limited anticoagulation without drug-related adverse effects in a phase 1 trial. When put in context with earlier observations suggesting that FXI contributes to venous thromboembolism and cardiovascular disease, although contributing minimally to hemostasis, our data further justify clinical evaluation of AB023 in conditions where contact-initiated FXI activation is suspected to have a pathogenic role. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03097341.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Factor XI/antagonistas & inhibidores , Factor XIa/fisiología , Fibrinolíticos/uso terapéutico , Adulto , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticoagulantes/efectos adversos , Anticoagulantes/inmunología , Anticoagulantes/farmacología , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Factor XI/inmunología , Factor XIIa/fisiología , Fibrinolíticos/efectos adversos , Fibrinolíticos/inmunología , Fibrinolíticos/farmacología , Oclusión de Injerto Vascular/tratamiento farmacológico , Humanos , Papio , Tiempo de Tromboplastina Parcial
7.
Metab Brain Dis ; 30(1): 57-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24810631

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by demyelination and axonal damage of the central nervous system. The pathogenesis of MS has also been linked to vascular inflammation and local activation of the coagulation system, resulting in perivascular fibrin deposition. Treatment of experimental autoimmune encephalomyelitis (EAE), a model of human MS, with antithrombotic and antiinflammatory activated protein C (APC) reduces disease severity. Since recombinant APC (Drotecogin alfa), originally approved for the treatment of severe sepsis, is not available for human MS studies, we tested the hypothesis that pharmacologic activation of endogenous protein C could likewise improve the outcome of EAE. Mice were immunized with murine myelin oligodendrocyte glycoprotein (MOG) peptides and at the onset of EAE symptoms, were treated every other day with either WE thrombin (25 µg/kg; i.v.), a selective recombinant protein C activator thrombin analog, or saline control. Mice were monitored for changes in disease score until euthanized for ex vivo analysis of inflammation. Administration of WE thrombin significantly ameliorated clinical severity of EAE, reduced inflammatory cell infiltration and demyelination, suppressed the activation of macrophages comprising the CD11b + population and reduced accumulation of fibrin (ogen) in the spinal cord. These data suggest that symptomatic MS may respond to a treatment strategy that involves temporal pharmacological enhancement of endogenous APC generation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteína C/agonistas , Trombina/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Activación Enzimática , Fibrina/análisis , Fibrinógeno/análisis , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Activación de Macrófagos , Masculino , Ratones , Esclerosis Múltiple , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Mutación Puntual , Proteína C/metabolismo , Médula Espinal/patología , Bazo/inmunología , Bazo/patología , Trombina/genética , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/biosíntesis , Sustancia Blanca/patología
8.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L508-20, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24381131

RESUMEN

Adiponectin is an adipose-derived hormone with anti-inflammatory activity. Following subacute ozone exposure (0.3 ppm for 24-72 h), neutrophilic inflammation and IL-6 are augmented in adiponectin-deficient (Adipo(-/-)) mice. The IL-17/granulocyte colony-stimulating factor (G-CSF) axis is required for this increased neutrophilia. We hypothesized that elevated IL-6 in Adipo(-/-) mice contributes to their augmented responses to ozone via effects on IL-17A expression. Therefore, we generated mice deficient in both adiponectin and IL-6 (Adipo(-/-)/IL-6(-/-)) and exposed them to ozone or air. In ozone-exposed mice, bronchoalveolar lavage (BAL) neutrophils, IL-6, and G-CSF, and pulmonary Il17a mRNA expression were greater in Adipo(-/-) vs. wild-type mice, but reduced in Adipo(-/-)/IL-6(-/-) vs. Adipo(-/-) mice. IL-17A(+) F4/80(+) cells and IL-17A(+) γδ T cells were also reduced in Adipo(-/-)/IL-6(-/-) vs. Adipo(-/-) mice exposed to ozone. Only BAL neutrophils were reduced in IL-6(-/-) vs. wild-type mice. In wild-type mice, IL-6 was expressed in Gr-1(+)F4/80(-)CD11c(-) cells, whereas in Adipo(-/-) mice F4/80(+)CD11c(+) cells also expressed IL-6, suggesting that IL-6 is regulated by adiponectin in these alveolar macrophages. Transcriptomic analysis identified serum amyloid A3 (Saa3), which promotes IL-17A expression, as the gene most differentially augmented by ozone in Adipo(-/-) vs. wild-type mice. After ozone, Saa3 mRNA expression was markedly greater in Adipo(-/-) vs. wild-type mice but reduced in Adipo(-/-)/IL-6(-/-) vs. Adipo(-/-) mice. In conclusion, our data support a pivotal role of IL-6 in the hyperinflammatory condition observed in Adipo(-/-) mice after ozone exposure and suggest that this role of IL-6 involves its ability to induce Saa3, IL-17A, and G-CSF.


Asunto(s)
Adiponectina/deficiencia , Inflamación/inmunología , Interleucina-6/metabolismo , Macrófagos Alveolares/metabolismo , Ozono/farmacología , Animales , Líquido del Lavado Bronquioalveolar/citología , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-6/genética , Pulmón/metabolismo , Recuento de Linfocitos , Macrófagos Alveolares/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Oxidantes Fotoquímicos/farmacología , ARN Mensajero/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Proteína Amiloide A Sérica/genética , Linfocitos T/citología
9.
Blood ; 119(20): 4762-8, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22442348

RESUMEN

Severe bacterial sepsis often leads to a systemic procoagulant and proinflammatory condition that can manifest as disseminated intravascular coagulation, septic shock, and multiple organ failure. Because activation of the contact proteases factor XII (FXII), prekallikrein, and factor XI (FXI) can trigger coagulation and inflammatory responses, the contact factors have been considered potential targets for the treatment of sepsis. However, the pathogenic role of contact activation in severe infections has not been well defined. We therefore investigated whether an anticoagulant antibody (14E11) that selectively inhibits prothrombotic FXI activation by activated FXII (FXIIa) modifies the course of bowel perforation-induced peritoneal sepsis in mice. Early anticoagulation with 14E11 suppressed systemic thrombin- antithrombin complex formation, IL-6, and TNF-α levels, and reduced platelet consumption in the circulation and deposition in the blood vessels. Treatment with 14E11 within 12 hours after bowel perforation significantly improved survival compared with vehicle treatment, and the saturating dose did not increase tail bleeding. These data suggest that severe polymicrobial abdominal infection induces prothrombotic FXI activation, to the detriment of the host. Systemic anticoagulation by inhibiting FXI activation or FXIIa procoagulant activity during sepsis may therefore limit the development of disseminated intravascular coagulation without increasing bleeding risks.


Asunto(s)
Anticoagulantes/farmacología , Trastornos de la Coagulación Sanguínea/prevención & control , Coinfección/tratamiento farmacológico , Coinfección/mortalidad , Inflamación/prevención & control , Sepsis/tratamiento farmacológico , Sepsis/mortalidad , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticoagulantes/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/mortalidad , Coinfección/complicaciones , Coinfección/patología , Regulación hacia Abajo/efectos de los fármacos , Factor XIa/antagonistas & inhibidores , Factor XIa/inmunología , Inmunoterapia , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína C/farmacología , Proteína C/uso terapéutico , Sepsis/complicaciones , Sepsis/patología , Análisis de Supervivencia
10.
J Immunol ; 188(9): 4558-67, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22474022

RESUMEN

Pulmonary responses to ozone, a common air pollutant, are augmented in obese individuals. Adiponectin, an adipose-derived hormone that declines in obesity, has regulatory effects on the immune system. To determine the role of adiponectin in the pulmonary inflammation induced by extended (48-72 h) low-dose (0.3 parts per million) exposure to ozone, adiponectin-deficient (Adipo(-/-)) and wild-type mice were exposed to ozone or to room air. In wild-type mice, ozone exposure increased total bronchoalveolar lavage (BAL) adiponectin. Ozone-induced lung inflammation, including increases in BAL neutrophils, protein (an index of lung injury), IL-6, keratinocyte-derived chemokine, LPS-induced CXC chemokine, and G-CSF were augmented in Adipo(-/-) versus wild-type mice. Ozone also increased IL-17A mRNA expression to a greater extent in Adipo(-/-) versus wild-type mice. Moreover, compared with control Ab, anti-IL-17A Ab attenuated ozone-induced increases in BAL neutrophils and G-CSF in Adipo(-/-) but not in wild-type mice, suggesting that IL-17A, by promoting G-CSF release, contributed to augmented neutrophilia in Adipo(-/-) mice. Flow cytometric analysis of lung cells revealed that the number of CD45(+)/F4/80(+)/IL-17A(+) macrophages and γδ T cells expressing IL-17A increased after ozone exposure in wild-type mice and further increased in Adipo(-/-) mice. The IL-17(+) macrophages were CD11c(-) (interstitial macrophages), whereas CD11c(+) macrophages (alveolar macrophages) did not express IL-17A. Taken together, the data are consistent with the hypothesis that adiponectin protects against neutrophil recruitment induced by extended low-dose ozone exposure by inhibiting the induction and/or recruitment of IL-17A in interstitial macrophages and/or γδ T cells.


Asunto(s)
Adiponectina/inmunología , Interleucina-17/inmunología , Macrófagos Alveolares/inmunología , Neutrófilos/inmunología , Oxidantes Fotoquímicos/efectos adversos , Ozono/efectos adversos , Neumonía/inmunología , Adiponectina/genética , Adiponectina/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Lavado Broncoalveolar , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Interleucina-17/genética , Interleucina-17/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/patología , Oxidantes Fotoquímicos/farmacología , Ozono/farmacología , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/metabolismo , Neumonía/patología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
11.
Res Sq ; 2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37131631

RESUMEN

Objective: Relapses in patients with relapsing-remitting multiple sclerosis (RRMS) are typically treated with high-dose corticosteroids including methylprednisolone. However, high-dose corticosteroids are associated with significant adverse effects, can increase the risk for other morbidities, and often do not impact disease course. Multiple mechanisms are proposed to contribute to acute relapses in RRMS patients, including neuroinflammation, fibrin formation and compromised blood vessel barrier function. The protein C activator, E-WE thrombin is a recombinant therapeutic in clinical development for its antithrombotic and cytoprotective properties, including protection of endothelial cell barrier function. In mice, treatment with E-WE thrombin reduced neuroinflammation and extracellular fibrin formation in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). We therefore tested the hypothesis that E-WE thrombin could reduce disease severity in a relapsing-remitting model of EAE. Methods: Female SJL mice were inoculated with proteolipid protein (PLP) peptide and treated with E-WE thrombin (25 µg/kg; iv) or vehicle at onset of detectable disease. In other experiments, E-WE thrombin was compared to methylprednisolone (100 mg/kg; iv) or the combination of both. Results: Compared to vehicle, administration of E-WE thrombin significantly improved disease severity of the initial attack and relapse and delayed onset of relapse as effectively as methylprednisolone. Both methylprednisolone and E-WE thrombin reduced demyelination and immune cell recruitment, and the combination of both treatments had an additive effect. Conclusion: The data presented herein demonstrate that E-WE thrombin is protective in mice with relapsing-remitting EAE, a widely used model of MS. Our data indicate that E-WE thrombin is as effective as high-dose methylprednisolone in improving disease score and may exert additional benefit when administered in combination. Taken together, these data suggest that E-WE thrombin may be an effective alternative to high-dose methylprednisolone for managing acute MS attacks.

12.
Handb Exp Pharmacol ; (208): 403-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22222708

RESUMEN

In this chapter, we will review what is known about muscarinic regulation of immune cells and the contribution of immune cell muscarinic receptors to inflammatory disease and immunity. In particular, immune cell expression of cholinergic machinery, muscarinic receptor subtypes and functional consequences of agonist stimulation will be reviewed. Lastly, this chapter will discuss the potential therapeutic effects of selective antagonists on immune cell function and inflammatory disease in recent animal studies and human clinical trials.


Asunto(s)
Antiinflamatorios/farmacología , Sistema Inmunológico/efectos de los fármacos , Inflamación/tratamiento farmacológico , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Receptores Muscarínicos/efectos de los fármacos , Acetilcolina/metabolismo , Animales , Humanos , Sistema Inmunológico/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Receptores Muscarínicos/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Am J Respir Cell Mol Biol ; 43(4): 487-97, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19915153

RESUMEN

Obese mice have increased responses to acute ozone (O(3)) exposure. T-cadherin is a binding protein for the high-molecular weight isoforms of adiponectin, an anti-inflammatory hormone that declines in obesity. The objective of the present study was to determine whether adiponectin affects pulmonary responses to O(3), and whether these effects are mediated through T-cadherin. We performed bronchoalveolar lavage (BAL) and measured pulmonary responsiveness to methacholine after acute air or O(3) exposure (2 ppm for 3 h) in adiponectin-deficient (Adipo(-/-)) or T-cadherin-deficient (T-Cad(-/-)) mice. O(3) increased pulmonary responses to methacholine and increased BAL neutrophils and protein to a greater extent in wild-type than in Adipo(-/-) mice, whereas T-cadherin deficiency had no effect. O(3)-induced increases in BAL IL-6 and keratinocyte-derived chemokine (KC), which contribute to O(3)-induced pulmonary neutrophilia, were also greater in wild-type than in Adipo(-/-) mice. In contrast, responses to O(3) were not altered by transgenic overexpression of adiponectin. To determine which adiponectin isoforms are present in the lung, Western blotting was performed. The hexameric isoform of adiponectin dominated in serum, whereas BAL was dominated by the high-molecular weight isoform of adiponectin. Interestingly, serum adiponectin was greater in T-Cad(-/-) versus wild-type mice, whereas BAL adiponectin was lower in T-Cad(-/-) versus wild-type mice, suggesting that T-cadherin may be important for transit of high-molecular weight adiponectin from the blood to the lung. Our results indicate that adiponectin deficiency inhibits pulmonary inflammation induced by acute O(3) exposure, and that T-cadherin does not mediate the effects of adiponectin responsible for these events.


Asunto(s)
Adiponectina/deficiencia , Adiponectina/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ozono/administración & dosificación , Ozono/farmacología , Adiponectina/sangre , Animales , Peso Corporal/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/citología , Cadherinas/deficiencia , Cadherinas/metabolismo , Recuento de Células , Citocinas/metabolismo , Exposición por Inhalación , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L340-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19465517

RESUMEN

Inhibitory M(2) muscarinic receptors on airway parasympathetic nerves normally limit acetylcholine release. Viral infections decrease M(2) receptor function, increasing vagally mediated bronchoconstriction. Since retinoic acid deficiency causes M(2) receptor dysfunction, we tested whether retinoic acid would prevent virus-induced airway hyperreactivity and prevent M(2) receptor dysfunction. Guinea pigs infected with parainfluenza virus were hyperreactive to electrical stimulation of the vagus nerves, but not to intravenous acetylcholine, indicating that hyperreactivity was due to increased release of acetylcholine from parasympathetic nerves. The muscarinic agonist pilocarpine, which inhibits vagally mediated bronchoconstriction in control animals, no longer inhibited vagally induced bronchoconstriction, demonstrating M(2) receptor dysfunction. Treatment with all-trans retinoic acid (1 mg/kg) prevented virus-induced hyperreactivity and M(2) receptor dysfunction. However, retinoic acid also significantly reduced viral titers in the lungs and attenuated virus-induced lung inflammation. In vitro, retinoic acid decreased M(2) receptor mRNA expression in both human neuroblastoma cells and primary cultures of airway parasympathetic neurons. Thus, the protective effects of retinoic acid on airway function during viral infection appear to be due to anti-inflammatory and antiviral mechanisms, rather than to direct effects on M(2) receptor gene expression.


Asunto(s)
Antineoplásicos/farmacología , Hiperreactividad Bronquial , Infecciones por Paramyxoviridae , Receptor Muscarínico M2/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Tretinoina/farmacología , Acetilcolina/farmacología , Animales , Hiperreactividad Bronquial/tratamiento farmacológico , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/virología , Broncoconstricción/efectos de los fármacos , Broncoconstricción/inmunología , Línea Celular Tumoral , Colinérgicos/farmacología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Femenino , Expresión Génica/efectos de los fármacos , Cobayas , Humanos , Interleucina-8/genética , Agonistas Muscarínicos/farmacología , Neuroblastoma , Neuronas/efectos de los fármacos , Neuronas/fisiología , Infecciones por Paramyxoviridae/complicaciones , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/inmunología , Pilocarpina/farmacología , Receptor Muscarínico M2/genética , Receptores Inmunológicos , Mucosa Respiratoria/citología , Mucosa Respiratoria/inervación , Tráquea/citología , Tráquea/efectos de los fármacos , Tráquea/inervación , Replicación Viral/efectos de los fármacos
15.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L228-37, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19447892

RESUMEN

Although anticholinergic therapy inhibits bronchoconstriction in asthmatic patients and antigen-challenged animals, administration of atropine 1 h before antigen challenge significantly potentiates airway hyperreactivity and eosinophil activation measured 24 h later. This potentiation in airway hyperreactivity is related to increased eosinophil activation and is mediated at the level of the airway nerves. Since eosinophils produce nerve growth factor (NGF), which is known to play a role in antigen-induced airway hyperreactivity, we tested whether NGF mediates atropine-enhanced, antigen challenge-induced hyperreactivity. Antibody to NGF (Ab NGF) was administered to sensitized guinea pigs with and without atropine pretreatment (1 mg/kg iv) 1 h before challenge. At 24 h after challenge, animals were anesthetized, vagotomized, paralyzed, and ventilated. Electrical stimulation of both vagus nerves caused bronchoconstriction that was increased in challenged animals. Atropine pretreatment potentiated antigen challenge-induced hyperreactivity. Ab NGF did not affect eosinophils or inflammatory cells in any group, nor did it prevent hyperreactivity in challenged animals that were not pretreated with atropine. However, Ab NGF did prevent atropine-enhanced, antigen challenge-induced hyperreactivity and eosinophil activation (assessed by immunohistochemistry). This effect was specific to NGF, since animals given control IgG remained hyperreactive. These data suggest that anticholinergic therapy amplifies eosinophil interactions with airway nerves via NGF. Therefore, therapeutic strategies that target both eosinophil activation and NGF-mediated inflammatory processes in allergic asthma are likely to be beneficial.


Asunto(s)
Antígenos/farmacología , Atropina/farmacología , Hiperreactividad Bronquial/inmunología , Broncodilatadores/farmacología , Eosinófilos/inmunología , Factor de Crecimiento Nervioso/metabolismo , Animales , Antígenos/inmunología , Asma/inducido químicamente , Asma/inmunología , Asma/fisiopatología , Presión Sanguínea/efectos de los fármacos , Bradicardia/inducido químicamente , Bradicardia/inmunología , Bradicardia/fisiopatología , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/fisiopatología , Broncoconstricción/efectos de los fármacos , Broncoconstricción/inmunología , Estimulación Eléctrica , Femenino , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/inmunología , Factor de Crecimiento Nervioso/inmunología , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Sistema Nervioso Parasimpático/inmunología , Receptor Muscarínico M2/fisiología , Organismos Libres de Patógenos Específicos , Vagotomía , Nervio Vago/fisiología
17.
J Allergy (Cairo) ; 2013: 349520, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861690

RESUMEN

Obesity is an important risk factor for asthma. Obese individuals have decreased circulating adiponectin, an adipose-derived hormone with anti-inflammatory properties. We hypothesized that transgenic overexpression of adiponectin would attenuate allergic airways inflammation and mucous hyperplasia in mice. To test this hypothesis, we used mice overexpressing adiponectin (Adipo Tg). Adipo Tg mice had marked increases in both serum adiponectin and bronchoalveolar lavage (BAL) fluid adiponectin. Both acute and chronic ovalbumin (OVA) sensitization and challenge protocols were used. In both protocols, OVA-induced increases in total BAL cells were attenuated in Adipo Tg versus WT mice. In the acute protocol, OVA-induced increases in several IL-13 dependent genes were attenuated in Adipo Tg versus WT mice, even though IL-13 per se was not affected. With chronic exposure, though OVA-induced increases in goblet cells numbers per millimeter of basement membrane were greater in Adipo Tg versus WT mice, mRNA abundance of mucous genes in lungs was not different. Also, adiponectin overexpression did not induce M2 polarization in alveolar macrophages. Our results indicate that adiponectin protects against allergen-induced inflammatory cell recruitment to the airspaces, but not development of goblet cell hyperplasia.

18.
Transl Stroke Res ; 3(3): 381-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23634198

RESUMEN

Blood coagulation factor XI (FXI) is an established risk factor for acute ischemic stroke (AIS) and thrombosis, but is also needed for normal hemostasis. Contact factor XII (FXII), an upstream activator of FXI, also contributes to experimental stroke, but is not required for hemostasis. We investigated whether selectively inhibiting FXII-mediated FXI activation, while leaving other FXI and FXII functions intact, could improve the outcome of experimental AIS in mice. Twenty-four hours before induction of AIS by placement of a filament into the internal carotid artery for 60 min, mice were anticoagulated with an antibody directed against the apple 2 domain of FXI. This antibody selectively reduces the prothrombotic activation of FXI by FXIIa but does not affect activated FXI or hemostatic activation of FXI by thrombin, thus leaving hemostasis intact in mice and primates. In this model of AIS/reperfusion injury, mice that received the antibody before AIS displayed less ischemic damage, manifested as reduced cerebral infarction and fibrin deposition (thrombosis), increased cortical reperfusion, and improved neurological behavior. Further, the antibody-anticoagulated mice had no detectable hemostasis impairment. Consistent with the neuroprotective phenotype of FXII-deficient mice, our data suggest that a single molecular event, FXII-mediated FXI activation, contributes to the development of experimental AIS.

19.
PLoS One ; 7(7): e41088, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815927

RESUMEN

Adiponectin is an adipose derived hormone that declines in obesity. We have previously shown that exogenous administration of adiponectin reduces allergic airways responses in mice. T-cadherin (T-cad; Cdh13) is a binding protein for the high molecular weight isoforms of adiponectin. To determine whether the beneficial effects of adiponectin on allergic airways responses require T-cad, we sensitized wildtype (WT), T-cadherin deficient (T-cad(-/-)) and adiponectin and T-cad bideficient mice to ovalbumin (OVA) and challenged the mice with aerosolized OVA or PBS. Compared to WT, T-cad(-/-) mice were protected against OVA-induced airway hyperresponsiveness, increases in BAL inflammatory cells, and induction of IL-13, IL-17, and eotaxin expression. Histological analysis of the lungs of OVA-challenged T-cad(-/-) versus WT mice indicated reduced inflammation around the airways, and reduced mucous cell hyperplasia. Combined adiponectin and T-cad deficiency reversed the effects of T-cad deficiency alone, indicating that the observed effects of T-cad deficiency require adiponectin. Compared to WT, serum adiponectin was markedly increased in T-cad(-/-) mice, likely because adiponectin that is normally sequestered by endothelial T-cad remains free in the circulation. In conclusion, T-cad does not mediate the protective effects of adiponectin. Instead, mice lacking T-cad have reduced allergic airways disease, likely because elevated serum adiponectin levels act on other adiponectin signaling pathways.


Asunto(s)
Adiponectina/metabolismo , Cadherinas/fisiología , Adiponectina/sangre , Animales , Hiperreactividad Bronquial/metabolismo , Lavado Broncoalveolar , Cadherinas/metabolismo , Cruzamientos Genéticos , Inflamación , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/farmacología , Unión Proteica , Transducción de Señal
20.
J Leukoc Biol ; 90(5): 923-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21807745

RESUMEN

PMN leukocytes are the most abundant leukocytes in the circulation and play an important role in host defense. PMN leukocyte recruitment and inflammatory responses at sites of infection are critical components in innate immunity. Although inflammation and coagulation are known to have bidirectional relationships, little is known about the interaction between PMN leukocytes and coagulation factors. Coagulation FXI participates in the intrinsic coagulation pathway upon its activation, contributing to hemostasis and thrombosis. We have shown previously that FXI-deficient mice have an increased survival and less leukocyte accumulation into the peritoneum in severe polymicrobial peritonitis. This result suggests a role for FXI in leukocyte trafficking and/or function. In this study, we characterized the functional consequences of FXIa binding to PMN leukocytes. FXIa reduced PMN leukocyte chemotaxis triggered by the chemokine, IL-8, or the bacterial-derived peptide, fMLP, perhaps as a result of the loss of directed migration. In summary, our data suggest that FXIa modulates the inflammatory response of PMN leukocytes by altering migration. These studies highlight the interplay between inflammation and coagulation and suggest that FXIa may play a role in innate immunity.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Factor XIa/metabolismo , Neutrófilos/fisiología , Coagulación Sanguínea , Movimiento Celular , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Factor XIa/farmacología , Humanos , Inflamación/metabolismo , Interleucina-8/farmacología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Peritonitis/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda