Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(11): e0294438, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37983208

RESUMO

BCR-ABL tyrosine kinase inhibitors (TKIs) have dramatically improved survival in Philadelphia chromosome-positive leukemias. Newer BCR-ABL TKIs provide superior cancer outcomes but with increased risk of acute arterial thrombosis, which further increases in patients with cardiovascular comorbidities and mitigates survival benefits compared to imatinib. Recent studies implicate endothelial cell (EC) damage in this toxicity by unknown mechanisms with few side-by-side comparisons of multiple TKIs and with no available data on endothelial impact of recently approved TKIs or novels TKIs being tested in clinical trials. To characterize BCR-ABL TKI induced EC dysfunction we exposed primary human umbilical vein ECs in 2D and 3D culture to clinically relevant concentrations of seven BCR-ABL TKIs and quantified their impact on EC scratch-wound healing, viability, inflammation, and permeability mechanisms. Dasatinib, ponatinib, and nilotinib, the TKIs associated with thrombosis in patients, all significantly impaired EC wound healing, survival, and proliferation compared to imatinib, but only dasatinib and ponatinib impaired cell migration and only nilotinib enhanced EC necrosis. Dasatinib and ponatinib increased leukocyte adhesion to ECs with upregulation of adhesion molecule expression in ECs (ICAM1, VCAM1, and P-selectin) and leukocytes (PSGL1). Dasatinib increased permeability and impaired cell junctional integrity in human engineered microvessels, consistent with its unique association with pleural effusions. Of the new agents, bafetinib decreased EC viability and increased microvessel permeability while asciminib and radotinib did not impact any EC function tested. In summary, the vasculotoxic TKIs (dasatinib, ponatinib, nilotinib) cause EC toxicity but with mechanistic differences, supporting the potential need for drug-specific vasculoprotective strategies. Asciminib and radotinib do not induce EC toxicity at clinically relevant concentrations suggesting a better safety profile.


Assuntos
Antineoplásicos , Leucemia Mielogênica Crônica BCR-ABL Positiva , Trombose , Humanos , Mesilato de Imatinib/efeitos adversos , Dasatinibe/efeitos adversos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Inibidores de Proteínas Quinases/toxicidade , Células Endoteliais , Trombose/tratamento farmacológico , Proteínas de Fusão bcr-abl , Antineoplásicos/uso terapêutico
3.
Curr Oncol Rep ; 24(4): 463-474, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35179707

RESUMO

PURPOSE OF REVIEW: While vascular endothelial growth factor receptor inhibitors (VEGFRis) have dramatically improved cancer survival, these drugs cause hypertension in a majority of patients. This side effect is often dose limiting and increases cardiovascular mortality in cancer survivors. This review summarizes recent advances in our understanding of the molecular mechanisms and clinical findings that impact management of VEGFRi-induced hypertension. RECENT FINDINGS: Recent studies define new connections between endothelial dysfunction and VEGFRi-induced hypertension, including the balance between nitric oxide, oxidative stress, endothelin signaling, and prostaglandins and the potential role of microparticles, vascular smooth muscle cells, vascular stiffness, and microvessel rarefaction. Data implicating genetic polymorphisms that might identify patients at risk for VEGFRi-induced hypertension and the growing body of literature associating VEGFRi-induced hypertension with antitumor efficacy are reviewed. These recent advances have implications for the future of cardio-oncology clinics and the management of VEGFRi-induced hypertension.


Assuntos
Hipertensão , Fator A de Crescimento do Endotélio Vascular , Inibidores da Angiogênese/efeitos adversos , Humanos , Hipertensão/induzido quimicamente , Hipertensão/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular , Transdução de Sinais
4.
Arterioscler Thromb Vasc Biol ; 40(8): 1838-1853, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32460581

RESUMO

OBJECTIVE: Vascular calcification is a cardiovascular risk factor and accelerated in diabetes mellitus. Previous work has established a role for calcification-prone extracellular vesicles in promoting vascular calcification. However, the mechanisms by which diabetes mellitus provokes cardiovascular events remain incompletely understood. Our goal was to identify that increased S100A9 promotes the release of calcification-prone extracellular vesicles from human macrophages in diabetes mellitus. Approach and Results: Human primary macrophages exposed to high glucose (25 mmol/L) increased S100A9 secretion and the expression of receptor for advanced glycation end products (RAGE) protein. Recombinant S100A9 induced the expression of proinflammatory and osteogenic factors, as well as the number of extracellular vesicles with high calcific potential (alkaline phosphatase activity, P<0.001) in macrophages. Treatment with a RAGE antagonist or silencing with S100A9 siRNA in macrophages abolished these responses, suggesting that stimulation of the S100A9-RAGE axis by hyperglycemia favors a procalcific environment. We further showed that an imbalance between Nrf-2 (nuclear factor 2 erythroid related factor 2) and NF-κB (nuclear factor-κB) pathways contributes to macrophage activation and promotes a procalcific environment. In addition, streptozotocin-induced diabetic Apoe-/-S100a9-/- mice and mice treated with S100a9 siRNA encapsulated in macrophage-targeted lipid nanoparticles showed decreased inflammation and microcalcification in atherosclerotic plaques, as gauged by molecular imaging and comprehensive histological analysis. In human carotid plaques, comparative proteomics in patients with diabetes mellitus and histological analysis showed that the S100A9-RAGE axis associates with osteogenic activity and the formation of microcalcification. CONCLUSIONS: Under hyperglycemic conditions, macrophages release calcific extracellular vesicles through mechanisms involving the S100A9-RAGE axis, thus contributing to the formation of microcalcification within atherosclerotic plaques.


Assuntos
Calgranulina B/fisiologia , Complicações do Diabetes/etiologia , Vesículas Extracelulares/fisiologia , Macrófagos/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Calcificação Vascular/etiologia , Animais , Diabetes Mellitus Experimental/complicações , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Placa Aterosclerótica/etiologia
6.
Blood ; 128(13): 1766-76, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27338096

RESUMO

The complement system plays a key role in innate immunity, inflammation, and coagulation. The system is delicately balanced by negative regulatory mechanisms that modulate the host response to pathogen invasion and injury. The serpin, C1-esterase inhibitor (C1-INH), is the only known plasma inhibitor of C1s, the initiating serine protease of the classical pathway of complement. Like other serpin-protease partners, C1-INH interaction with C1s is accelerated by polyanions such as heparin. Polyphosphate (polyP) is a naturally occurring polyanion with effects on coagulation and complement. We recently found that polyP binds to C1-INH, prompting us to consider whether polyP acts as a cofactor for C1-INH interactions with its target proteases. We show that polyP dampens C1s-mediated activation of the classical pathway in a polymer length- and concentration-dependent manner by accelerating C1-INH neutralization of C1s cleavage of C4 and C2. PolyP significantly increases the rate of interaction between C1s and C1-INH, to an extent comparable to heparin, with an exosite on the serine protease domain of the enzyme playing a major role in this interaction. In a serum-based cell culture system, polyP significantly suppressed C4d deposition on endothelial cells, generated via the classical and lectin pathways. Moreover, polyP and C1-INH colocalize in activated platelets, suggesting that their interactions are physiologically relevant. In summary, like heparin, polyP is a naturally occurring cofactor for the C1s:C1-INH interaction and thus an important regulator of complement activation. The findings may provide novel insights into mechanisms underlying inflammatory diseases and the development of new therapies.


Assuntos
Proteínas Inativadoras do Complemento 1/metabolismo , Proteínas do Sistema Complemento/metabolismo , Polifosfatos/metabolismo , Sítios de Ligação , Plaquetas/imunologia , Plaquetas/metabolismo , Células Cultivadas , Proteína Inibidora do Complemento C1 , Complemento C1s/química , Complemento C1s/metabolismo , Complemento C2/metabolismo , Complemento C4/metabolismo , Via Clássica do Complemento , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Heparina/metabolismo , Humanos , Técnicas In Vitro , Polifosfatos/química
7.
Blood Adv ; 1(1): 62-74, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-29296696

RESUMO

Heparin-induced thrombocytopenia (HIT) is a thrombotic disorder initiated by antibodies to complexes between platelet factor 4 (PF4) and heparin. The risk of recurrent thromboembolism persists after heparin is cleared and platelet activation leading to release of PF4 has dissipated. We asked whether antigenic complexes between polyphosphates and PF4 released from activated platelets might intensify or sustain the prothrombotic phenotype of HIT. PF4 forms stable, ultralarge complexes with polyphosphates of various sizes, including those released from platelets, which are recognized by the HIT-like monoclonal KKO, an immunoglobulin G2bκ monoclonal heparin/PF4 binding antibody, and by human HIT antibodies. KKO helps to protect PF4/polyphosphate complexes from degradation by phosphatases. Complement is activated when HIT antibodies bind to PF4/polyphosphate complexes and PF4 reverses the inhibition of complement by polyphosphates. Polyphosphates and PF4 are stored primarily in separate granules in resting platelets, but they colocalize when the cells are activated. Platelets activated by subaggregating doses of thrombin receptor activating peptide release polyphosphates and PF4, which form antigenic complexes that allow KKO to further activate platelets in the absence of heparin and exogenous PF4. These studies suggest that thrombin- or immune complex-mediated release of endogenous antigenic PF4/polyphosphate complexes from platelets may augment the prothrombotic risk of HIT and perpetuate the risk of thrombosis after heparin has been discontinued.

8.
Blood ; 126(12): 1494-502, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26136249

RESUMO

Factor XIIa (FXIIa) and factor XIa (FXIa) contribute to thrombosis in animal models, whereas platelet-derived polyphosphate (polyP) may potentiate contact or thrombin-feedback pathways. The significance of these mediators in human blood under thrombotic flow conditions on tissue factor (TF) -bearing surfaces remains inadequately resolved. Human blood (corn trypsin inhibitor treated [4 µg/mL]) was tested by microfluidic assay for clotting on collagen/TF at TF surface concentration ([TF]wall) from ∼0.1 to 2 molecules per µm(2). Anti-FXI antibodies (14E11 and O1A6) or polyP-binding protein (PPXbd) were used to block FXIIa-dependent FXI activation, FXIa-dependent factor IX (FIX) activation, or platelet-derived polyP, respectively. Fibrin formation was sensitive to 14E11 at 0 to 0.1 molecules per µm(2) and sensitive to O1A6 at 0 to 0.2 molecules per µm(2). However, neither antibody reduced fibrin generation at ∼2 molecules per µm(2) when the extrinsic pathway became dominant. Interestingly, PPXbd reduced fibrin generation at low [TF]wall (0.1 molecules per µm(2)) but not at zero or high [TF]wall, suggesting a role for polyP distinct from FXIIa activation and requiring low extrinsic pathway participation. Regardless of [TF]wall, PPXbd enhanced fibrin sensitivity to tissue plasminogen activator and promoted clot retraction during fibrinolysis concomitant with an observed PPXbd-mediated reduction of fibrin fiber diameter. This is the first detection of endogenous polyP function in human blood under thrombotic flow conditions. When triggered by low [TF]wall, thrombosis may be druggable by contact pathway inhibition, although thrombolytic susceptibility may benefit from polyP antagonism regardless of [TF]wall.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fator XIa/metabolismo , Polifosfatos/metabolismo , Tromboplastina/metabolismo , Anticorpos/farmacologia , Testes de Coagulação Sanguínea , Plaquetas/citologia , Colágeno/metabolismo , Humanos , Terapia de Alvo Molecular , Transdução de Sinais , Trombina/metabolismo , Trombose/tratamento farmacológico , Trombose/metabolismo
9.
Int J Rheum Dis ; 18(5): 587-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26082352

RESUMO

AIM: Sir Benjamin Collins Brodie (1783-1862) was a London surgeon who investigated joint disease by observation and morbid anatomy in the first half of the 19th century. His descriptions of inflammatory joint disease have been referred to in the past, but only in a fragmentary way. This study aimed to provide more detail than previously, allowing for a new appreciation of his contributions. METHODS: The authors used the first (1818), third (1834) and fifth (1850) editions of his book, Pathological and Surgical Observations on the Diseases of Joints, to provide his description of reactive arthritis, rheumatoid arthritis and ankylosing spondylitis. RESULTS: Brodie's descriptions are admirably clear. He describes reactive arthritis in the first edition of his book more completely than authors before him and even Reiter a century later. He recognised that the conjunctivo-urethral-synovial syndrome can occur independently of gonorrhoea, that there are often repeated attacks, and that irits is a complication - the first indication that this syndrome might be part of what we now call seronegative spondyloarthritis. The fifth edition (1850) has among the earliest descriptions in English of ankylosing spondylitis and rheumatoid arthritis. CONCLUSION: Brodie's understanding of rheumatic diseases, and his willingness to pass this knowledge on, demands that he be widely recognised as a pioneer rheumatologist.


Assuntos
Reumatologia/história , Inglaterra , História do Século XIX , Londres
10.
Crit Rev Biochem Mol Biol ; 50(4): 326-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26018600

RESUMO

The plasma coagulation system in mammalian blood consists of a cascade of enzyme activation events in which serine proteases activate the proteins (proenzymes and procofactors) in the next step of the cascade via limited proteolysis. The ultimate outcome is the polymerization of fibrin and the activation of platelets, leading to a blood clot. This process is protective, as it prevents excessive blood loss following injury (normal hemostasis). Unfortunately, the blood clotting system can also lead to unwanted blood clots inside blood vessels (pathologic thrombosis), which is a leading cause of disability and death in the developed world. There are two main mechanisms for triggering the blood clotting, termed the tissue factor pathway and the contact pathway. Only one of these pathways (the tissue factor pathway) functions in normal hemostasis. Both pathways, however, are thought to contribute to thrombosis. An emerging concept is that the contact pathway functions in host pathogen defenses. This review focuses on how the initiation phase of the blood clotting cascade is regulated in both pathways, with a discussion of the contributions of these pathways to hemostasis versus thrombosis.


Assuntos
Coagulação Sanguínea , Modelos Biológicos , Tromboplastina/metabolismo , Animais , Hemostasia , Humanos , Ativação Plaquetária , Proteólise , Tromboplastina/análise , Trombose/sangue , Trombose/metabolismo
11.
Sci Transl Med ; 6(260): 260ra150, 2014 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-25355700

RESUMO

Heparin-based anticoagulant drugs have been widely used for the prevention of blood clotting during surgical procedures and for the treatment of thromboembolic events. However, bleeding risks associated with these anticoagulants demand continuous monitoring and neutralization with suitable antidotes. Protamine, the only clinically approved antidote to heparin, has shown adverse effects and ineffectiveness against low-molecular weight heparins and fondaparinux, a heparin-related medication. Alternative approaches based on cationic molecules and recombinant proteins have several drawbacks including limited efficacy, toxicity, immunogenicity, and high cost. Thus, there is an unmet clinical need for safer, rapid, predictable, and cost-effective anticoagulant-reversal agents for all clinically used heparins. We report a design strategy for a fully synthetic dendritic polymer-based universal heparin reversal agent (UHRA) that makes use of multivalent presentation of branched cationic heparin binding groups (HBGs). Optimization of the UHRA design was aided by isothermal titration calorimetry studies, biocompatibility evaluation, and heparin neutralization analysis. By controlling the scaffold's molecular weight, the nature of the protective shell, and the presentation of HBGs on the polymer scaffold, we arrived at lead UHRA molecules that completely neutralized the activity of all clinically used heparins. The optimized UHRA molecules demonstrated superior efficacy and safety profiles and mitigated heparin-induced bleeding in animal models. This new polymer therapeutic may benefit patients undergoing high-risk surgical procedures and has potential for the treatment of anticoagulant-related bleeding problems.


Assuntos
Anticoagulantes/síntese química , Heparina/síntese química , Anticoagulantes/farmacologia , Calorimetria , Heparina/farmacologia
12.
Blood ; 124(22): 3183-90, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25202141

RESUMO

Polyphosphate (polyP) is secreted by activated platelets and has been shown to contribute to thrombosis, suggesting that it could be a novel antithrombotic target. Previously reported polyP inhibitors based on polycationic substances, such as polyethylenimine, polyamidoamine dendrimers, and polymyxin B, although they attenuate thrombosis, all have significant toxicity in vivo, likely due to the presence of multiple primary amines responsible for their polyP binding ability. In this study, we examined a novel class of nontoxic polycationic compounds initially designed as universal heparin reversal agents (UHRAs) to determine their ability to block polyP procoagulant activity and also to determine their utility as antithrombotic treatments. Several UHRA compounds strongly inhibited polyP procoagulant activity in vitro, and 4 were selected for further examination in mouse models of thrombosis and hemostasis. Compounds UHRA-9 and UHRA-10 significantly reduced arterial thrombosis in mice. In mouse tail bleeding tests, administration of UHRA-9 or UHRA-10 was associated with significantly less bleeding compared with therapeutically equivalent doses of heparin. Thus, these compounds offer a new platform for developing novel antithrombotic agents that target procoagulant anionic polymers such as polyP with reduced toxicity and bleeding side effects.


Assuntos
Dendrímeros/farmacologia , Fibrinolíticos/farmacologia , Hemostasia/efeitos dos fármacos , Polifosfatos/antagonistas & inibidores , Trombose/prevenção & controle , Animais , Coagulação Sanguínea/efeitos dos fármacos , Dendrímeros/efeitos adversos , Dendrímeros/química , Fibrinolíticos/efeitos adversos , Fibrinolíticos/química , Heparina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Polifosfatos/metabolismo , Ligação Proteica/efeitos dos fármacos , Trombina/metabolismo , Trombose/sangue
13.
Blood ; 120(26): 5103-10, 2012 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22968458

RESUMO

Inorganic polyphosphates are linear polymers of orthophosphate that modulate blood clotting and inflammation. Polyphosphate accumulates in infectious microorganisms and is secreted by activated platelets; long-chain polyphosphate in particular is an extremely potent initiator of the contact pathway, a limb of the clotting cascade important for thrombosis but dispensable for hemostasis. Polyphosphate inhibitors therefore might act as novel antithrombotic/anti-inflammatory agents with reduced bleeding side effects. Antipolyphosphate antibodies are unlikely because of polyphosphate's ubiquity and simple structure; and although phosphatases such as alkaline phosphatase can digest polyphosphate, they take time and may degrade other biologically active molecules. We now identify a panel of polyphosphate inhibitors, including cationic proteins, polymers, and small molecules, and report their effectiveness in vitro and in vivo. We also compare their effectiveness against the procoagulant activity of RNA. Polyphosphate inhibitors were antithrombotic in mouse models of venous and arterial thrombosis and blocked the inflammatory effect of polyphosphate injected intradermally in mice. This study provides proof of principle for polyphosphate inhibitors as antithrombotic/anti-inflammatory agents in vitro and in vivo, with a novel mode of action compared with conventional anticoagulants.


Assuntos
Anti-Inflamatórios/farmacologia , Fibrinolíticos/farmacologia , Inflamação/tratamento farmacológico , Polifosfatos/antagonistas & inibidores , Trombose/tratamento farmacológico , Animais , Anti-Inflamatórios/isolamento & purificação , Coagulação Sanguínea/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Fibrinolíticos/isolamento & purificação , Hemostasia/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Inflamação/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Polifosfatos/sangue , Trombose/sangue
14.
Blood ; 120(22): 4421-31, 2012 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22955924

RESUMO

Soluble immune complexes (ICs) are abundant in autoimmune diseases, yet neutrophil responses to these soluble humoral factors remain uncharacterized. Moreover, the individual role of the uniquely human FcγRIIA and glycophosphatidylinositol (GPI)-linked FcγRIIIB in IC-mediated inflammation is still debated. Here we exploited mice and cell lines expressing these human neutrophil FcγRs to demonstrate that FcγRIIIB alone, in the absence of its known signaling partners FcγRIIA and the integrin Mac-1, internalizes soluble ICs through a mechanism used by GPI-anchored receptors and fluid-phase endocytosis. FcγRIIA also uses this pathway. As shown by intravital microscopy, FcγRIIA but not FcγRIIIB-mediated neutrophil interactions with extravascular soluble ICs results in the formation of neutrophil extracellular traps (NETs) in tissues. Unexpectedly, in wild-type mice, IC-induced NETosis does not rely on the NADPH oxidase, myeloperoxidase, or neutrophil elastase. In the context of soluble ICs present primarily within vessels, FcγRIIIB-mediated neutrophil recruitment requires Mac-1 and is associated with the removal of intravascular IC deposits. Collectively, our studies assign a new role for FcγRIIIB in the removal of soluble ICs within the vasculature that may serve to maintain homeostasis, whereas FcγRIIA engagement of tissue soluble ICs generates NETs, a proinflammatory process linked to autoimmunity.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Endocitose , Neutrófilos/metabolismo , Receptores de IgG/fisiologia , Animais , Autoimunidade/genética , Endocitose/fisiologia , Espaço Extracelular/imunologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Solubilidade , Transfecção , Regulação para Cima/imunologia , Regulação para Cima/fisiologia
15.
J Immunol ; 188(3): 1421-30, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22219321

RESUMO

T cell subset-specific migration to inflammatory sites is tightly regulated and involves interaction of the T cells with the endothelium. Th17 cells often appear at different inflammatory sites than Th1 cells, or both subsets appear at the same sites but at different times. Differences in T cell subset adhesion to endothelium may contribute to subset-specific migratory behavior, but this possibility has not been well studied. We examined the adhesion of mouse Th17 cells to endothelial adhesion molecules and endothelium under flow in vitro and to microvessels in vivo and we characterized their migratory phenotype by flow cytometry and quantitative RT-PCR. More Th17 than Th1 cells interacted with E-selectin. Fewer Th17 than Th1 cells bound to TNF-α-activated E-selectin-deficient endothelial cells, and intravital microscopy studies demonstrated that Th17 cells engage in more rolling interactions with TNF-α-treated microvessels than Th1 cells in wild-type mice but not in E-selectin-deficient mice. Th17 adhesion to ICAM-1 was dependent on integrin activation by CCL20, the ligand for CCR6, which is highly expressed by Th17 cells. In an air pouch model of inflammation, CCL20 triggered recruitment of Th17 but not Th1 cells. These data provide evidence that E-selectin- and ICAM-1-dependent adhesion of Th17 and Th1 cells with endothelium are quantitatively different.


Assuntos
Adesão Celular/imunologia , Endotélio Vascular/imunologia , Células Th1/fisiologia , Animais , Quimiocina CCL20/metabolismo , Selectina E/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Células Th17
16.
Arterioscler Thromb Vasc Biol ; 31(11): 2464-72, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21903941

RESUMO

OBJECTIVE: Evidence has linked collagen loss with the onset of acute coronary events. This study tested the hypothesis that selective matrix metalloproteinase-13 (MMP-13) collagenase inhibition increases collagen content in already established and nascent mouse atheromas. METHODS AND RESULTS: In vitro and in situ experiments documented the selectivity and efficacy of an orally available MMP-13 inhibitor (MMP13i-A). In vivo observations monitored macrophage accumulation and MMP-13 activity using molecular imaging. After 10 weeks of MMP13i-A treatment, apolipoprotein E-deficient mice with evolving or established lesions exhibited reduced MMP-13 activity without affecting macrophage content, measured either by intravital microscopy or fluorescence reflectance imaging. Histological analysis indicated that MMP13-iA did not affect plaque size or macrophage or smooth muscle cell accumulation. Administration of MMP13i-A to mice with evolving or established atheromas substantially increased plaque interstitial collagen content in the intima and locally in the fibrous cap, compared with vehicle-treated controls. Analysis of collagen revealed thicker collagen fibers within the plaques of treated groups. CONCLUSION: Pharmacological MMP-13 inhibition yields collagen accumulation in plaques (a feature associated in humans with resistance to rupture), even in established plaques. This study, of considerable clinical relevance, furnishes new mechanistic insight into regulation of the plaque's extracellular matrix and validates molecular imaging for studying plaque biology.


Assuntos
Aterosclerose/metabolismo , Colágeno/metabolismo , Inibidores Enzimáticos/farmacologia , Metaloproteinase 13 da Matriz/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/patologia , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Modelos Animais de Doenças , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
18.
J Law Med ; 12(2): 178-87, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15575320

RESUMO

The medical indemnity crisis in Australia forced doctors, lawyers and insurers to re-appraise the way they handle claims for compensation for medical error. This article examines some of the new approaches available in Australia when patients claim compensation from their doctors.


Assuntos
Compensação e Reparação/legislação & jurisprudência , Dissidências e Disputas/legislação & jurisprudência , Revisão da Utilização de Seguros/legislação & jurisprudência , Austrália , Humanos , Advogados/legislação & jurisprudência , Erros Médicos/legislação & jurisprudência , Médicos/legislação & jurisprudência
19.
J Clin Rheumatol ; 9(4): 228-38, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17041463

RESUMO

This report describes a visit by an international group interested in Rheumatology to the Rheumatology centers and traditional Chinese medicine units in the People's Republic of China. Differing disease patterns and treatment approaches offer opportunities for studies and collaborations. We can also learn from the traditional Chinese approach with individualization of therapy and attention to health maintenance.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...