Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34782457

RESUMO

Lipofuscin granules enclose mixtures of cross-linked proteins and lipids in proportions that depend on the tissue analyzed. Retinal lipofuscin is unique in that it contains mostly lipids with very little proteins. However, retinal lipofuscin also presents biological and physicochemical characteristics indistinguishable from conventional granules, including indigestibility, tendency to cause lysosome swelling that results in rupture or defective functions, and ability to trigger NLRP3 inflammation, a symptom of low-level disruption of lysosomes. In addition, like conventional lipofuscins, it appears as an autofluorescent pigment, considered toxic waste, and a biomarker of aging. Ocular lipofuscin accumulates in the retinal pigment epithelium (RPE), whereby it interferes with the support of the neuroretina. RPE cell death is the primary cause of blindness in the most prevalent incurable genetic and age-related human disorders, Stargardt disease and age-related macular degeneration (AMD), respectively. Although retinal lipofuscin is directly linked to the cell death of the RPE in Stargardt, the extent to which it contributes to AMD is a matter of debate. Nonetheless, the number of AMD clinical trials that target lipofuscin formation speaks for the potential relevance for AMD as well. Here, we show that retinal lipofuscin triggers an atypical necroptotic cascade, amenable to pharmacological intervention. This pathway is distinct from canonic necroptosis and is instead dependent on the destabilization of lysosomes. We also provide evidence that necroptosis is activated in aged human retinas with AMD. Overall, this cytotoxicity mechanism may offer therapeutic targets and markers for genetic and age-related diseases associated with lipofuscin buildups.


Assuntos
Membranas Intracelulares/metabolismo , Lipofuscina/farmacologia , Lisossomos/metabolismo , Necroptose/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Envelhecimento , Oxirredutases do Álcool , Animais , Morte Celular , Humanos , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
2.
Blood ; 137(16): 2221-2230, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33512476

RESUMO

Reduced plasma fibrinolysis has been identified as a potential risk factor for venous thromboembolism (VTE), but the role of cell surface fibrinolysis in VTE is unknown. The annexin A2/S100A10 complex serves as a coreceptor for plasminogen and tissue plasminogen activator (tPA), augmenting plasmin generation by 60-fold on the endothelial cell surface. Several studies in both mice and humans support the concept that A2 regulates fibrin homeostasis and intravascular thrombosis in vivo. Here, we examined A2 protein expression and function in 115 adult subjects with VTE and 87 healthy controls. Using peripheral blood mononuclear cells as a surrogate for endothelial cells, we found a 41% mean decrease in cell surface tPA-dependent fibrinolytic activity in subjects who had a positive personal and family history of VTE but tested negative for known inherited thrombophilias (ITs). A2 protein was reduced on average by 70% and messenger RNA levels by 30%, but neither decrease correlated with anticoagulant therapy. Neither cell A2 protein nor cell surface plasmin generation correlated with plasma-based clot lysis times, suggesting that the plasma and cell surface fibrinolytic systems operate independently of one another. These data suggest that reduced expression of annexin A2 protein is associated with cell surface hypofibrinolysis and may represent a novel risk factor for IT.


Assuntos
Anexina A2/genética , Fibrinólise , Tromboembolia Venosa/genética , Adulto , Regulação para Baixo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , Tromboembolia Venosa/sangue , Adulto Jovem
3.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34202091

RESUMO

As a cell surface tissue plasminogen activator (tPA)-plasminogen receptor, the annexin A2 (A2) complex facilitates plasmin generation on the endothelial cell surface, and is an established regulator of hemostasis. Whereas A2 is overexpressed in hemorrhagic disease such as acute promyelocytic leukemia, its underexpression or impairment may result in thrombosis, as in antiphospholipid syndrome, venous thromboembolism, or atherosclerosis. Within immune response cells, A2 orchestrates membrane repair, vesicle fusion, and cytoskeletal organization, thus playing a critical role in inflammatory response and tissue injury. Dysregulation of A2 is evident in multiple human disorders, and may contribute to the pathogenesis of various inflammatory disorders. The fibrinolytic system, moreover, is central to wound healing through its ability to remodel the provisional matrix and promote angiogenesis. A2 dysfunction may also promote tissue fibrogenesis and end-organ fibrosis.


Assuntos
Anexina A2/genética , Suscetibilidade a Doenças , Fibrinólise/genética , Fibrose/etiologia , Inflamação/etiologia , Animais , Anexina A2/metabolismo , Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Biomarcadores , Fibrose/metabolismo , Hemostasia/genética , Humanos , Imunidade , Inflamação/metabolismo , Especificidade de Órgãos , Regeneração
4.
J Immunol ; 197(4): 1252-61, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27371724

RESUMO

Cryptococcus neoformans is a fungal pathogen with a unique intracellular pathogenic strategy that includes nonlytic exocytosis, a phenomenon whereby fungal cells are expunged from macrophages without lysing the host cell. The exact mechanism and specific proteins involved in this process have yet to be completely defined. Using murine macrophages deficient in the membrane phospholipid binding protein, annexin A2 (ANXA2), we observed a significant decrease in both phagocytosis of yeast cells and the frequency of nonlytic exocytosis. Cryptococcal cells isolated from Anxa2-deficient (Anxa2(-/-)) bone marrow-derived macrophages and lung parenchyma displayed significantly larger capsules than those isolated from wild-type macrophages and tissues. Concomitantly, we observed significant differences in the amount of reactive oxygen species produced between Anxa2(-/-) and Anxa2(+/+) macrophages. Despite comparable fungal burden, Anxa2(-/-) mice died more rapidly than wild-type mice when infected with C. neoformans, and Anxa2(-/-) mice exhibited enhanced inflammatory responses, suggesting that the reduced survival reflected greater immune-mediated damage. Together, these findings suggest a role for ANXA2 in the control of cryptococcal infection, macrophage function, and fungal morphology.


Assuntos
Anexina A2/imunologia , Criptococose/imunologia , Cryptococcus neoformans/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Animais , Anexina A2/metabolismo , Cryptococcus neoformans/patogenicidade , Modelos Animais de Doenças , Exocitose/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Virulência
5.
J Cell Sci ; 127(Pt 4): 828-44, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24357721

RESUMO

The annexins are an evolutionarily conserved family of phospholipid-binding proteins of largely unknown function. We observed that the AnxA2(-/-) lung basement membrane specifically lacks collagen VI (COL6), and postulated that ANXA2 directs bronchial epithelial cell secretion of COL6, an unusually large multimeric protein. COL6 serves to anchor cells to basement membranes and, unlike other collagens, undergoes multimerization prior to secretion. Here, we show that AnxA2(-/-) mice have reduced exercise tolerance with impaired lung tissue elasticity, which was phenocopied in Col6a1(-/-) mice. In vitro, AnxA2(-/-) fibroblasts retained COL6 within intracellular vesicles and adhered poorly to their matrix unless ANXA2 expression was restored. In vivo, AnxA2(-/-) bronchial epithelial cells underwent apoptosis and disadhesion. Immunoprecipitation and immunoelectron microscopy revealed that ANXA2 associates with COL6 and the SNARE proteins SNAP-23 and VAMP2 at secretory vesicle membranes of bronchial epithelial cells, and that absence of ANXA2 leads to retention of COL6 in a late-Golgi, VAMP2-positive compartment. These results define a new role for ANXA2 in the COL6 secretion pathway, and further show that this pathway establishes cell-matrix interactions that underlie normal pulmonary function and epithelial cell survival.


Assuntos
Anexina A2/fisiologia , Apoptose , Colágeno Tipo VI/metabolismo , Células Epiteliais/metabolismo , Animais , Membrana Basal , Brônquios/metabolismo , Brônquios/patologia , Forma Celular , Sobrevivência Celular , Células Cultivadas , Colágeno Tipo VI/genética , Elasticidade , Células Epiteliais/fisiologia , Tolerância ao Exercício , Complexo de Golgi/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esforço Físico , Transporte Proteico , Ventilação Pulmonar , Mucosa Respiratória/patologia , Proteína 2 Associada à Membrana da Vesícula/metabolismo
6.
Biol Chem ; 397(10): 1005-16, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27366903

RESUMO

The formation of new blood vessels from pre-existing vasculature, the process known as angiogenesis, is highly regulated by pro- and anti-angiogenic signaling molecules including growth factors and proteases. As an endothelial cell-surface co-receptor for plasminogen and tissue plasminogen activator, the annexin A2 (ANXA2) complex accelerates plasmin generation and facilitates fibrinolysis. Plasmin can subsequently activate a downstream proteolytic cascade involving multiple matrix metalloproteinases. Thus, in addition to maintaining blood vessel patency, the ANXA2 complex can also promote angiogenesis via its pro-fibrinolytic activity. The generation of ANXA2-deficient mice allowed us to first observe the pro-angiogenic role of ANXA2 in vivo. Further investigations have provided additional details regarding the mechanism for ANXA2 regulation of retinal and corneal angiogenesis. Other studies have reported that ANXA2 supports angiogenesis in specific tumor-related settings. Here, we summarize results from in vivo studies that illustrate the pro-angiogenic role of ANXA2, and discuss the critical questions that may lead to an advanced understanding of the molecular mechanisms for ANXA2-mediated angiogenesis. Finally, highlights from studies on ANXA2-interacting agents offer potential therapeutic opportunities for the application of ANXA2-centered pharmaceuticals in angiogenesis-related disorders.


Assuntos
Anexina A2/metabolismo , Neovascularização Fisiológica , Animais , Fibrinólise , Humanos , Neoplasias/irrigação sanguínea , Neovascularização Patológica
7.
Blood ; 121(26): 5192-202, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23632889

RESUMO

Pericytes and vascular smooth muscle cells (VSMCs), which are recruited to developing blood vessels by platelet-derived growth factor BB, support endothelial cell survival and vascular stability. Here, we report that imatinib, a tyrosine kinase inhibitor of platelet-derived growth factor receptor ß (PDGFRß), impaired growth of lymphoma in both human xenograft and murine allograft models. Lymphoma cells themselves neither expressed PDGFRß nor were growth inhibited by imatinib. Tumor growth inhibition was associated with decreased microvascular density and increased vascular leakage. In vivo, imatinib induced apoptosis of tumor-associated PDGFRß(+) pericytes and loss of perivascular integrity. In vitro, imatinib inhibited PDGFRß(+) VSMC proliferation and PDGF-BB signaling, whereas small interfering RNA knockdown of PDGFRß in pericytes protected them against imatinib-mediated growth inhibition. Fluorescence-activated cell sorter analysis of tumor tissue revealed depletion of pericytes, endothelial cells, and their progenitors following imatinib treatment. Compared with imatinib, treatment with an anti-PDGFRß monoclonal antibody partially inhibited lymphoma growth. Last, microarray analysis (Gene Expression Omnibus database accession number GSE30752) of PDGFRß(+) VSMCs following imatinib treatment showed down-regulation of genes implicated in vascular cell proliferation, survival, and assembly, including those representing multiple pathways downstream of PDGFRß. Taken together, these data indicate that PDGFRß(+) pericytes may represent a novel, nonendothelial, antiangiogenic target for lymphoma therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Benzamidas/farmacologia , Linfoma Difuso de Grandes Células B/prevenção & controle , Linfoma de Células T/prevenção & controle , Neovascularização Patológica/prevenção & controle , Pericitos/efeitos dos fármacos , Piperazinas/farmacologia , Pirimidinas/farmacologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Diferenciação Celular , Proliferação de Células , Perfilação da Expressão Gênica , Humanos , Mesilato de Imatinib , Técnicas Imunoenzimáticas , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Pericitos/imunologia , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas
8.
Trans Am Clin Climatol Assoc ; 126: 144-55, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26330668

RESUMO

Annexin A2 is a multicompartmental protein that orchestrates a spectrum of dynamic membrane-related events. At cell surfaces, A2 forms the (A2•S100A10)2 complex which accelerates tissue plasminogen activator-dependent activation of the fibrinolytic protease, plasmin. Anti-A2 antibodies are associated with clinical thrombosis in antiphospholipid syndrome, whereas overexpression of A2 promotes hyperfibrinolytic bleeding in acute promyelocytic leukemia. A2 is upregulated in hypoxic tissues, and mice deficient in A2 are resistant to hypoxia-related retinal neovascularization in a model of diabetic retinopathy. Within the cell, A2 regulates membrane fusion processes involved in the secretion of pre-packaged, ultra-large molecules. In stimulated dendritic cells, A2 maintains lysosomal membrane integrity, thereby modulating inflammasome activation and cytokine secretion. Together, these findings suggest an emerging, multifaceted role for annexin A2 in human health and disease. The author's work has been inspired by numerous colleagues and mentors, and by the author's grandfather, and former ACCA member, Dr. J. Burns Amberson.


Assuntos
Anexina A2/metabolismo , Fibrinólise , Imunidade Inata , Animais , Membrana Celular/metabolismo , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Humanos , Neovascularização Patológica , Transdução de Sinais
9.
Genesis ; 52(7): 657-70, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24740971

RESUMO

Epidermal growth factor-like domain 7 (Egfl7) expression in the developing embryo is largely restricted to sites of mesodermal progenitors of angioblasts/hemangioblasts and the vascular endothelium. We hypothesize that Egfl7 marks the endothelial lineage during embryonic development, and can be used to define the emergence of endothelial progenitor cells, as well as to visualize newly-forming vasculature in the embryo and during the processes of physiologic and pathologic angiogenesis in the adult. We have generated a transgenic mouse strain that expresses enhanced green fluorescent protein (eGFP) under the control of a minimal Egfl7 regulatory sequence (Egfl7:eGFP). Expression of the transgene recapitulated that of endogenous Egfl7 at sites of vasculogenesis and angiogenesis in the allantois, yolk sac, and in the embryo proper. The transgene was not expressed in the quiescent endothelium of most adult organs. However, the uterus and ovary, which undergo vascular growth and remodeling throughout the estrus cycle, expressed high levels of Egfl7:eGFP. Importantly, expression of the Egfl7:eGFP transgene was induced in adult neovasculature. We also found that increased Egfl7 expression contributed to pathologic revascularization in the mouse retina. To our knowledge, this is the first mouse model that enables monitoring of endothelial cells at sites of active vasculogenesis and angiogenesis. This model also facilitated the isolation and characterization of EGFL7(+) endothelial cell populations by fluorescence activated cell sorting (FACS). Together, our results demonstrate that the Egfl7:eGFP reporter mouse is a valuable tool that can be used to elucidate the mechanisms by which blood vessels form during development and under pathologic circumstances.


Assuntos
Linhagem da Célula , Células Progenitoras Endoteliais/metabolismo , Neovascularização Fisiológica , Proteínas/genética , Alantoide/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Família de Proteínas EGF , Células Progenitoras Endoteliais/citologia , Feminino , Camundongos , Neovascularização Patológica , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Proteínas/metabolismo , Vasos Retinianos/metabolismo , Retinopatia da Prematuridade/metabolismo , Útero/crescimento & desenvolvimento , Útero/metabolismo , Saco Vitelino/metabolismo
10.
Semin Thromb Hemost ; 39(4): 338-46, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23483454

RESUMO

Annexin A2 (A2) is a multicompartmental, multifunctional protein that orchestrates a growing spectrum of biologic processes. At the endothelial cell surface, A2 and S100A10 (p11) form a heterotetramer, which accelerates tissue plasminogen activator-dependent activation of the fibrinolytic protease, plasmin. In antiphospholipid syndrome, anti-A2 antibodies are associated with clinical thrombosis, whereas overexpression of A2 in acute promyelocytic leukemia promotes hyperfibrinolytic bleeding. A2 is upregulated in hypoxia, and mice deficient in A2 are resistant to oxygen-induced retinal neovascularization, suggesting a role for A2 in human retinal vascular proliferation. In solid malignancies, the (A2•p11)(2) tetramer may promote cancer cell invasion, whereas in multiple myeloma A2 enables malignant plasmacyte growth and predicts prognosis. In the central nervous system, the p11 enables membrane insertion of serotonin receptors that govern mood. In the peripheral nervous system, p11 directs sodium channels to the plasma membrane, enabling pain perception. In cerebral cortex neurons, A2 stabilizes the microtubule-associated tau protein, which, when mutated, is associated with frontotemporal dementia. In inflammatory dendritic cells, A2 maintains late endosomal/lysosomal membrane integrity, thus modulating inflammasome activation and cytokine secretion in a model of aseptic arthritis. Together, these findings suggest an emerging, multifaceted role for A2 in human health and disease.


Assuntos
Anexina A2/fisiologia , Animais , Anexina A2/metabolismo , Membrana Celular/metabolismo , Fibrinólise/fisiologia , Humanos , Proteínas S100/metabolismo
11.
Blood ; 118(10): 2918-29, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21788340

RESUMO

Oxygen-induced retinopathy (OIR) is a well-characterized model for retinopathy of prematurity, a disorder that results from rapid microvascular proliferation after exposure of the retina to high oxygen levels. Here, we report that the proliferative phase of OIR requires transcriptional induction of the annexin A2 (A2) gene through the direct action of the hypoxia-inducible factor-1 complex. We show, in addition, that A2 stabilizes its binding partner, p11, and promotes OIR-related angiogenesis by enabling clearance of perivascular fibrin. Adenoviral-mediated restoration of A2 expression restores neovascularization in the oxygen-primed Anxa2(-/-) retina and reinstates plasmin generation and directed migration in cultured Anxa2(-/-) endothelial cells. Systemic depletion of fibrin repairs the neovascular response to high oxygen treatment in the Anxa2(-/-) retina, whereas inhibition of plasminogen activation dampens angiogenesis under the same conditions. These findings show that the A2 system enables retinal neoangiogenesis in OIR by enhancing perivascular activation of plasmin and remodeling of fibrin. These data suggest new potential approaches to retinal angiogenic disorders on the basis of modulation of perivascular fibrinolysis.


Assuntos
Anexina A2/fisiologia , Fibrina/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Patológica , Oxigênio/efeitos adversos , Doenças Retinianas/etiologia , Doenças Retinianas/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Western Blotting , Movimento Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Ensaio de Imunoadsorção Enzimática , Fibrinolisina/metabolismo , Fibrinólise , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Técnicas Imunoenzimáticas , Imunoprecipitação , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativadores de Plasminogênio/metabolismo , Regiões Promotoras Genéticas/genética , Estabilidade de RNA , RNA Mensageiro/genética , Doenças Retinianas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
12.
Invest Ophthalmol Vis Sci ; 64(4): 33, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37103008

RESUMO

Purpose: We examined the role of annexin A2 (A2) in the development of diabetic retinal vasculopathy by testing the effect of Anxa2 gene deletion as well as administration of anti-A2 antibodies on pericyte dropout and retinal neovascularization in diabetic Akita mice, and in mice subjected to oxygen-induced retinopathy. Methods: We analyzed diabetic Ins2AKITA mice with or without global deletion of Anxa2, as well as Ins2AKITA mice that received intravitreal anti-A2 IgG or control antibody at 2, 4, and 6 months, for retinal pericyte dropout at 7 months of age. In addition, we assessed the effect of intravitreal anti-A2 on oxygen-induced retinopathy (OIR) in neonatal mice by quantifying retinal neovascular and vaso-obliterative area, and by enumeration of neovascular tufts. Results: Both deletion of the Anxa2 gene and immunologic blockade of A2 prevented pericyte depletion in retinas of diabetic Ins2AKITA mice. Blockade of A2 also reduced vaso-obliteration and neovascularization in the OIR model of vascular proliferation. This effect was amplified when a combination of antivascular endothelial growth factor (VEGF) and anti-A2 antibodies was used. Conclusions: Therapeutic approaches that target A2, alone or in combination with anti-VEGF therapy, are effective in mice, and may also curtail the progression of retinal vascular disease in humans with diabetes.


Assuntos
Anexina A2 , Diabetes Mellitus , Retinopatia Diabética , Doenças Retinianas , Neovascularização Retiniana , Animais , Camundongos , Anexina A2/genética , Anexina A2/metabolismo , Anexina A2/uso terapêutico , Retinopatia Diabética/prevenção & controle , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Oxigênio/toxicidade , Oxigênio/metabolismo , Doenças Retinianas/metabolismo , Neovascularização Retiniana/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Crit Care Explor ; 5(2): e0862, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36798534

RESUMO

The primary objective of this study was to determine whether expression of the multifunctional and adherens junction-regulating protein, annexin A2 (A2), is altered following cardiopulmonary bypass (CPB). A secondary objective was to determine whether depletion of A2 is associated with post-CPB organ dysfunction in children. DESIGN: In a prospective, observational study conducted over a 1-year period in children undergoing cardiac surgery requiring CPB, we analyzed A2 expression in peripheral blood mononuclear cells at different time points. We then assessed the relationship of A2 expression with organ function at each time point in the early postoperative period. SETTING: Twenty-three-bed mixed PICU in a tertiary academic center. PARTICIPANTS: Patients 1 month to 18 years old undergoing cardiac surgery requiring CPB. MEAN OUTCOME MEASUREMENTS AND RESULTS: We analyzed A2 expression in 22 enrolled subjects (n = 9, 1-23 mo old; n = 13, 2-18 yr old) and found a proteolysis-mediated decline in intact A2 immediately after bypass (p = 0.0009), reaching a median of 4% of baseline at 6 hours after bypass (p < 0.0001), and recovery by postoperative day 1. The degree of A2 depletion immediately after bypass in 1-23-month-olds correlated strongly with the extent of organ dysfunction, as measured by PICU admission Vasoactive-Ventilation-Renal (p = 0.004) and PEdiatric Logistic Organ Dysfunction-2 (p = 0.039) scores on postoperative day 1. A2 depletion immediately after bypass also correlated with more protracted requirement for both respiratory support (p = 0.007) and invasive ventilation (p = 0.013) in the 1-23-month-olds. CONCLUSIONS AND RELEVANCE: The degree of depletion of A2 following CPB correlates with more severe organ dysfunction, especially acute respiratory compromise in children under 2 years. These findings suggest that loss of A2 may contribute to pulmonary microvascular leak in young children following CPB.

14.
J Neurosci ; 31(40): 14346-60, 2011 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21976520

RESUMO

Gliomas are highly invasive, lethal brain tumors. Tumor-associated proteases play an important role in glioma progression. Annexin A2 is overexpressed in many cancers and correlates with increased plasmin activity on the tumor cell surface, which mediates degradation of extracellular matrix and promotes neoangiogenesis to facilitate tumor growth. In this study, we used two glioma cell lines, mouse GL261-EGFP and rat C6/LacZ, as well as stable clones transfected with an annexin A2 knockdown construct. We find that the annexin A2 knockdown decreased glioma cell migration in vitro and decreased membrane-bound plasmin activity. In vivo, we injected the glioma cells into the rodent brain and followed glioma progression. Knockdown of annexin A2 in glioma cells decreased tumor size and slowed tumor progression, as evidenced by decreased invasion, angiogenesis, and proliferation, as well as increased apoptosis in the tumor tissue of the annexin A2 knockdown group. Moreover, we report that the levels of expression of annexin A2 in human glioma samples correlate with their degree of malignancy. Together, our findings demonstrate that inhibition of annexin A2 expression in glioma cells could become a new target for glioma therapy.


Assuntos
Anexina A2/biossíntese , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Progressão da Doença , Glioma/metabolismo , Glioma/patologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica/patologia , Projetos Piloto , Ratos , Ratos Endogâmicos F344
15.
J Biol Chem ; 286(17): 15428-39, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21115493

RESUMO

In response to blood vessel injury, hemostasis is initiated by platelet activation, advanced by thrombin generation, and tempered by fibrinolysis. The primary fibrinolytic protease, plasmin, can be activated either on a fibrin-containing thrombus or on cells. Annexin A2 (A2) heterotetramer (A2·p11)(2) is a key profibrinolytic complex that assembles plasminogen and tissue plasminogen activator and promotes plasmin generation. We now report that, in endothelial cells, plasmin specifically induces activation of conventional PKC, which phosphorylates serine 11 and serine 25 of A2, triggering dissociation of the (A2·p11)(2) tetramer. The resulting free p11 undergoes ubiquitin-mediated proteasomal degradation, thus preventing further translocation of A2 to the cell surface. In vivo, pretreatment of A2(+/+) but not A2(-/-) mice with a conventional PKC inhibitor significantly reduced thrombosis in a carotid artery injury model. These results indicate that augmentation of fibrinolytic vascular surveillance by blockade of serine phosphorylation is A2-dependent. We also demonstrate that plasmin-induced phosphorylation of A2 requires both cleavage of A2 and activation of Toll-like receptor 4 on the cell surface. We propose that plasmin can limit its own generation by triggering a finely tuned "feedback" mechanism whereby A2 becomes serine-phosphorylated, dissociates from p11, and fails to translocate to the cell surface.


Assuntos
Anexina A2/metabolismo , Células Endoteliais/metabolismo , Retroalimentação Fisiológica , Fibrinolisina/biossíntese , Proteína Quinase C/metabolismo , Animais , Camundongos , Camundongos Knockout , Fosforilação , Multimerização Proteica , Transporte Proteico , Serina/metabolismo
16.
Gastroenterology ; 140(2): 583-595.e4, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20826156

RESUMO

BACKGROUND & AIMS: Prograstrin induces proliferation in colon crypts by activating p65nuclear factor-κB (NF-κB) (p65) and ß-catenin. We investigated whether Annexin A2 (AnxA2), a progastrin receptor, activates NF-κB and ß-catenin in vivo. METHODS: ANXA2-null (ANXA2(-/-)) and wild-type (ANXA2(+/+)) mice were studied, along with clones of progastrin-responsive HEK-293 cells that stably expressed full-length progastrin (HEK-mGAS) or an empty vector (HEK-C). Small interfering RNA was used to down-regulate AnxA2, p65NF-κB, and ß-catenin in cells. RESULTS: Proliferation and activation of p65 and ß-catenin increased significantly in HEK-mGAS compared with HEK-C clones. HEK-mGAS cells had a 2- to 4-fold increase in relative levels of c-Myc, cyclooxygenase (COX)-2, CyclinD1, double cortin CAM kinase-like 1 (DCAMKL+1), and CD44, compared with HEK-C clones. Down-regulation of AnxA2 in HEK-mGAS clones reduced activation of NF-κB and ß-catenin, as well as levels of DCAMKL+1. Surprisingly, down-regulation of ß-catenin had no effect on activation of p65NF-κB, whereas down-regulation of p65 significantly reduced activation of ß-catenin in HEK-mGAS clones. Loss of either p65 or ß-catenin significantly reduced proliferation of HEK-mGAS clones, indicating that both factors are required for the proliferative effects of progastrin. Lengths of colon crypts and levels of p65, ß-catenin, DCAMKL+1, and CD44 were significantly higher in ANXA2(+/+) mice compared with either ANXA2(-/-) mice given progastrin or ANXA2(+/+) and ANXA2(-/-) mice given saline. CONCLUSIONS: AnxA2 expression is required for the biologic effects of progastrin in vivo and in vitro and mediates the stimulatory effect of progastrin on p65NF-κ, ß-catenin, and the putative stem cell markers DCAMKL+1 and CD44. AnxA2 might therefore mediate the hyperproliferative and cocarcinogenic effects of progastrin.


Assuntos
Anexina A2/metabolismo , Gastrinas/metabolismo , NF-kappa B/metabolismo , Precursores de Proteínas/metabolismo , Células-Tronco/metabolismo , Regulação para Cima , beta Catenina/metabolismo , Animais , Anexina A2/genética , Proliferação de Células , Colo/química , Colo/metabolismo , Ciclina D1/análise , Ciclo-Oxigenase 2/análise , Quinases Semelhantes a Duplacortina , Regulação para Baixo , Gastrinas/farmacologia , Células HEK293 , Humanos , Receptores de Hialuronatos/análise , Peptídeos e Proteínas de Sinalização Intracelular/análise , Camundongos , Camundongos Endogâmicos C57BL , Precursores de Proteínas/farmacologia , Proteínas Serina-Treonina Quinases/análise , Proteínas Proto-Oncogênicas c-myc/análise , RNA Interferente Pequeno/metabolismo , Células-Tronco/efeitos dos fármacos
17.
J Biomed Biotechnol ; 2012: 406273, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23193360

RESUMO

Since its discovery as a src kinase substrate more than three decades ago, appreciation for the physiologic functions of annexin A2 and its associated proteins has increased dramatically. With its binding partner S100A10 (p11), A2 forms a cell surface complex that regulates generation of the primary fibrinolytic protease, plasmin, and is dynamically regulated in settings of hemostasis and thrombosis. In addition, the complex is transcriptionally upregulated in hypoxia and promotes pathologic neoangiogenesis in the tissues such as the retina. Dysregulation of both A2 and p11 has been reported in examples of rodent and human cancer. Intracellularly, A2 plays a critical role in endosomal repair in postarthroplastic osteolysis, and intracellular p11 regulates serotonin receptor activity in psychiatric mood disorders. In human studies, the A2 system contributes to the coagulopathy of acute promyelocytic leukemia, and is a target of high-titer autoantibodies in patients with antiphospholipid syndrome, cerebral thrombosis, and possibly preeclampsia. Polymorphisms in the human ANXA2 gene have been associated with stroke and avascular osteonecrosis of bone, two severe complications of sickle cell disease. Together, these new findings suggest that manipulation of the annexin A2/S100A10 system may offer promising new avenues for treatment of a spectrum of human disorders.


Assuntos
Anexina A2/metabolismo , Doença , Saúde , Proteínas S100/metabolismo , Animais , Modelos Animais de Doenças , Humanos
19.
Stroke ; 42(2): 501-3, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21193750

RESUMO

BACKGROUND AND PURPOSE: Cerebral venous thrombosis (CVT) may be a manifestation of underlying autoimmune disease. Antibodies against annexin A2 (anti-A2Ab) coincide with antiphospholipid syndrome, in which antiphospholipid antibodies (aPLA) are associated with thrombosis in any vascular bed. Annexin A2, a profibrinolytic receptor and binding site for ß2-glycoprotein-I, the main target for aPLA, is highly expressed on cerebral endothelium. Here we evaluate the prevalence of anti-A2Ab in CVT. METHODS: Forty individuals with objectively documented CVT (33 women and 7 men) and 145 healthy controls were prospectively studied for hereditary and acquired prothrombotic risk factors, classical aPLA, and anti-A2Ab. RESULTS: One or more prothrombotic risk factors were found in 85% of CVT subjects, (pregnancy/puerperium in 57.5%, classical aPLA in 22.5%, and hereditary procoagulant risk factors in 17.5%). Anti-A2Ab (titer >3 SD) were significantly more prevalent in patients with CVT (12.5%) than in healthy individuals (2.1%, P<0.01, OR, 5.9). CONCLUSIONS: Anti-A2Ab are significantly associated with CVT and may define a subset of individuals with immune-mediated cerebral thrombosis.


Assuntos
Anexina A2/imunologia , Autoanticorpos/biossíntese , Fibrinólise/imunologia , Trombose Intracraniana/imunologia , Trombose Venosa/imunologia , Adolescente , Adulto , Biomarcadores/sangue , Feminino , Humanos , Trombose Intracraniana/sangue , Trombose Intracraniana/diagnóstico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Trombose Venosa/sangue , Trombose Venosa/diagnóstico , Adulto Jovem
20.
Am J Physiol Heart Circ Physiol ; 301(2): H617-24, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21602468

RESUMO

Nitric oxide (NO) is an important vasoactive molecule produced by three NO synthase (NOS) enzymes: neuronal (nNOS), inducible (iNOS), and endothelial NOS (eNOS). While eNOS contributes to blood vessel dilation that protects against the development of hypertension, iNOS has been primarily implicated as a disease-promoting isoform during atherogenesis. Despite this, iNOS may play a physiological role via the modulation of cyclooxygenase and thromboregulatory eicosanoid production. Herein, we examined the role of iNOS in a murine model of thrombosis. Blood flow was measured in carotid arteries of male and female wild-type (WT) and iNOS-deficient mice following ferric chloride-induced thrombosis. Female WT mice were more resistant to thrombotic occlusion than male counterparts but became more susceptible upon iNOS deletion. In contrast, male mice (with and without iNOS deletion) were equally susceptible to thrombosis. Deletion of iNOS was not associated with a change in the balance of thromboxane A(2) (TxA(2)) or antithrombotic prostacyclin (PGI(2)). Compared with male counterparts, female WT mice exhibited increased urinary nitrite and nitrate levels and enhanced ex vivo induction of iNOS in hearts and aortas. Our findings suggest that iNOS-derived NO in female WT mice may attenuate the effects of vascular injury. Thus, although iNOS is detrimental during atherogenesis, physiological iNOS levels may contribute to providing protection against thrombotic occlusion, a phenomenon that may be enhanced in female mice.


Assuntos
Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Trombose/prevenção & controle , Análise de Variância , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Coagulação Sanguínea , Cardiomegalia/enzimologia , Cardiomegalia/fisiopatologia , Artérias Carótidas/fisiopatologia , Lesões das Artérias Carótidas/induzido quimicamente , Lesões das Artérias Carótidas/complicações , Lesões das Artérias Carótidas/fisiopatologia , Cloretos , Modelos Animais de Doenças , Estradiol/urina , Feminino , Compostos Férricos , Interferon gama/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/urina , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Fluxo Sanguíneo Regional , Fatores Sexuais , Trombose/enzimologia , Trombose/etiologia , Trombose/fisiopatologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa