Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
Artigo em Inglês | MEDLINE | ID: mdl-35534208

RESUMO

The formation of new blood and lymphatic vessels is essential for both the development of multicellular organisms and (patho)physiological processes like wound repair and tumor growth. In the 1990s, circulating blood platelets were first postulated to regulate tumor angiogenesis by interacting with the endothelium and releasing angiogenic regulators from specialized α granules. Since then, many studies have validated the contributions of platelets to tumor angiogenesis, while uncovering novel roles for platelets in other angiogenic processes like wound resolution and retinal vascular disease. Although the majority of (lymph)angiogenesis occurs during development, platelets appear necessary for lymphatic but not vascular growth, implying their particular importance in pathological cases of adult angiogenesis. Future work is required to determine whether drugs targeting platelet production or function offer a clinically relevant tool to limit detrimental angiogenesis.


Assuntos
Vasos Linfáticos , Neoplasias , Humanos , Plaquetas/fisiologia , Neovascularização Fisiológica , Neovascularização Patológica
3.
Sci Adv ; 8(41): eabo5224, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36223471

RESUMO

Despite abundant research demonstrating that platelets can promote tumor cell metastasis, whether primary tumors affect platelet-producing megakaryocytes remains understudied. In this study, we used a spontaneous murine model of breast cancer to show that tumor burden reduced megakaryocyte number and size and disrupted polyploidization. Single-cell RNA sequencing demonstrated that megakaryocytes from tumor-bearing mice exhibit a pro-inflammatory phenotype, epitomized by increased Ctsg, Lcn2, S100a8, and S100a9 transcripts. Protein S100A8/A9 and lipocalin-2 levels were also increased in platelets, suggesting that tumor-induced alterations to megakaryocytes are passed on to their platelet progeny, which promoted in vitro tumor cell invasion and tumor cell lung colonization to a greater extent than platelets from wild-type animals. Our study is the first to demonstrate breast cancer-induced alterations in megakaryocytes, leading to qualitative changes in platelet content that may feedback to promote tumor metastasis.


Assuntos
Megacariócitos , Neoplasias , Animais , Plaquetas/metabolismo , Catepsina G/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Lipocalina-2/metabolismo , Camundongos , Neoplasias/metabolismo
4.
Blood Adv ; 6(20): 5668-5675, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-35482455

RESUMO

Programmed death ligand 1 (PD-L1) is an immune checkpoint protein that suppresses cytotoxic T lymphocytes and is often overexpressed in cancers. Due to favorable clinical trial results, immune checkpoint inhibition (ICI) is part of Food and Drug Administration approved immuno-oncology therapies; however, not all patients benefit from ICI therapy. High blood platelet-to-lymphocyte ratio has been associated with failure of ICI treatment, but whether platelets have a role in hindering ICI response is unclear. Here, we report that coculturing platelets with cancer cell lines increased protein and gene expression of tumor cell PD-L1, which was reduced by antiplatelet agents, such as aspirin and ticagrelor. Platelet cytokine arrays revealed that the well-established cytokines, including interferon-γ, were not the main regulators of platelet-mediated PD-L1 upregulation. Instead, the high molecular weight epidermal growth factor (EGF) is abundant in platelets, which caused an upregulation of tumor cell PD-L1. Both an EGF-neutralizing antibody and cetuximab (EGF receptor [EGFR] monoclonal antibody) inhibited platelet-induced increases in tumor cell PD-L1, suggesting that platelets induce tumor cell PD-L1 in an EGFR-dependent manner. Our data reveal a novel mechanism for platelets in tumor immune escape and warrant further investigation to determine if targeting platelets improves ICI therapeutic responses.


Assuntos
Antígeno B7-H1 , Neoplasias , Estados Unidos , Humanos , Antígeno B7-H1/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Interferon gama/farmacologia , Plaquetas/metabolismo , Inibidores de Checkpoint Imunológico , Proteínas de Checkpoint Imunológico , Cetuximab , Inibidores da Agregação Plaquetária , Ticagrelor , Receptores ErbB/metabolismo , Neoplasias/tratamento farmacológico , Aspirina , Anticorpos Neutralizantes
5.
Hematol Oncol Clin North Am ; 35(6): 1069-1084, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34391603

RESUMO

Bleeding disorders due to platelet dysfunction are a common hematologic complication affecting patients, and typically present with mucocutaneous bleeding or hemorrhage. An inherited platelet disorder should be suspected in individuals with a suggestive family history and no identified secondary causes of bleeding. Genetic defects have been described at all levels of platelet activation, including receptor binding, signaling, granule release, cytoskeletal remodeling, and platelet hematopoiesis. Management of these disorders is typically supportive, with an emphasis on awareness, patient education, and anticipatory guidance to prevent future episodes of bleeding.


Assuntos
Transtornos Plaquetários , Transtornos da Coagulação Sanguínea , Transtornos Plaquetários/diagnóstico , Transtornos Plaquetários/genética , Transtornos Plaquetários/terapia , Plaquetas , Hemorragia/genética , Humanos , Ativação Plaquetária
6.
Blood ; 137(23): 3174-3180, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33940594

RESUMO

Platelets have long been known to play important roles beyond hemostasis and thrombosis. Now recognized as a bona fide mediator of malignant disease, platelets influence various aspects of cancer progression, most notably tumor cell metastasis. Interestingly, platelets isolated from cancer patients often display distinct RNA and protein profiles, with no clear alterations in hemostatic activity. This phenotypically distinct population, termed tumor-educated platelets, now receive significant attention for their potential use as a readily available liquid biopsy for early cancer detection. Although the mechanisms underpinning platelet education are still being defined, direct uptake and storage of tumor-derived factors, signal-dependent changes in platelet RNA processing, and differential platelet production by tumor-educated megakaryocytes are the most prominent scenarios. This article aims to cover the various modalities of platelet education by tumors, in addition to assessing their diagnostic potential.


Assuntos
Plaquetas/metabolismo , Megacariócitos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , RNA Neoplásico/metabolismo , Animais , Plaquetas/patologia , Humanos , Biópsia Líquida , Megacariócitos/patologia , Metástase Neoplásica , Neoplasias/diagnóstico , Neoplasias/patologia
7.
Oncologist ; 25(10): e1500-e1508, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32881209

RESUMO

Coronavirus disease 2019 (COVID-19) is a current global pandemic caused by the novel coronavirus SARS-CoV-2. Alongside its potential to cause severe respiratory illness, studies have reported a distinct COVID-19-associated coagulopathy that is characterized by elevated D-dimer levels, hyperfibrinogenemia, mild thrombocytopenia, and slight prolongation of the prothrombin time. Studies have also reported increased rates of thromboembolism in patients with COVID-19, but variations in study methodologies, patient populations, and anticoagulation strategies make it challenging to distill implications for clinical practice. Here, we present a practical review of current literature and uses a case-based format to discuss the diagnostic approach and management of COVID-19-associated coagulopathy. IMPLICATIONS FOR PRACTICE: Coronavirus disease 2019 (COVID-19)-associated coagulopathy is characterized by elevated D-dimer levels, hyperfibrinogenemia, and increased rates of thromboembolism. Current management guidelines are based on limited evidence from retrospective studies that should be interpreted carefully. At this time, all hospitalized patients with suspected or confirmed COVID-19 should receive coagulation test surveillance and standard doses of prophylactic anticoagulation until prospective randomized controlled trials yield definitive information in support of higher prophylactic doses.


Assuntos
COVID-19/complicações , Trombose/diagnóstico , Trombose/terapia , Anticoagulantes/uso terapêutico , Biomarcadores/sangue , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/diagnóstico , Transtornos da Coagulação Sanguínea/terapia , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Fibrinogênio/análise , Humanos , SARS-CoV-2 , Tromboembolia/sangue , Tromboembolia/diagnóstico , Tromboembolia/prevenção & controle , Trombose/sangue , Trombose/complicações
8.
Blood Adv ; 3(20): 3092-3098, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31648331

RESUMO

In addition to their primary roles in hemostasis and thrombosis, platelets participate in many other physiological and pathological processes, including, but not limited to inflammation, wound healing, tumor metastasis, and angiogenesis. Among their most interesting properties is the large number of bioactive proteins stored in their α-granules, the major storage granule of platelets. We previously showed that platelets differentially package pro- and antiangiogenic proteins in distinct α-granules that undergo differential release upon platelet activation. Nevertheless, how megakaryocytes achieve differential packaging is not fully understood. In this study, we use a mouse megakaryocyte culture system and endocytosis assay to establish when and where differential packaging occurs during platelet production. Live cell microscopy of primary mouse megakaryocytes incubated with fluorescently conjugated fibrinogen and endostatin showed differential endocytosis and packaging of the labeled proteins into distinct α-granule subpopulations. Super-resolution microscopy of mouse proplatelets and human whole-blood platelet α-granules simultaneously probed for 2 different membrane proteins (VAMP-3 and VAMP-8), and multiple granular content proteins (bFGF, ENDO, TSP, VEGF) confirmed differential packaging of protein contents into α-granules. These data suggest that megakaryocytes differentially sort and package α-granule contents, which are preserved as α-granule subpopulations during proplatelet extension and platelet production.


Assuntos
Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Megacariócitos/metabolismo , Animais , Transporte Biológico , Biomarcadores , Diferenciação Celular , Imunofluorescência , Humanos , Megacariócitos/citologia , Camundongos , Trombopoese
9.
J Natl Cancer Inst ; 111(7): 684-694, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30624689

RESUMO

BACKGROUND: Vitamins are among the most frequently used supplements (48% of US adults). However, little is known about contributions of genetic variation to their efficacy and safety. Multiple pathways link catechol-O-methyltransferase (COMT) to the vitamin E supplement, alpha-tocopherol, and cancer. METHODS: Here we determined if COMT exerted pharmacogenetic effects on cancer prevention in two randomized trials of alpha-tocopherol supplementation. Pharmacogenetic effects of common COMT rs4680 (val158met), which encodes a nonsynonymous valine-to-methionine substitution, were examined in the trial plus a 10-year post-trial follow-up (overall) period of The Women's Genome Health Study (WGHS, N = 23 294), a 10-year alpha-tocopherol and aspirin trial with 10 years post-trial follow-up. Results were validated in a case/control (N = 2396/2235) subset of the Alpha-Tocopherol Beta-Carotene Cancer Prevention Study (ATBC, N = 29 133). The primary outcome was total cancers. Rates of cancer types prevalent in women (colorectal, breast, lung, uterine, and lymphoma/leukemia) were also examined. All statistical tests were two-sided. RESULTS: Random-effects meta-analysis of rs4680 genotype strata, in WGHS and ATBC overall periods, revealed differential alpha-tocopherol effects compared with placebo: met/met (hazard ratio [HR] = 0.88; 95% confidence interval [CI] = 0.80 to 0.97; P = .01), val/met (HR = 0.99; 95% CI = 0.92 to 1.06; P = .74), and val/val (HR = 1.18; 95% CI = 1.06 to 1.31; P = .002) with a statistically significant COMT by alpha-tocopherol interaction (Pinteraction <.001). Timing of effects differed, with stronger effects in WGHS trial and ATBC post-trial. CONCLUSION: Pharmacogenetic analysis of COMT and cancer prevention in two large randomized trials revealed statistically significant COMT by alpha-tocopherol interaction, such that alpha-tocopherol was beneficial among rs4680 met-allele (28.0%), but not val-allele (22.8%) homozygotes. These effects indicate the need for additional studies of genetic variation as a determinant of the benefits and possible harms of over-the-counter supplements, like alpha-tocopherol, used for health promotion.


Assuntos
Catecol O-Metiltransferase/genética , Estudos de Associação Genética , Neoplasias/genética , alfa-Tocoferol/uso terapêutico , Alelos , Suplementos Nutricionais/efeitos adversos , Feminino , Genótipo , Humanos , Masculino , Neoplasias/dietoterapia , Neoplasias/patologia , Neoplasias/prevenção & controle , Testes Farmacogenômicos , Ensaios Clínicos Controlados Aleatórios como Assunto , alfa-Tocoferol/efeitos adversos , beta Caroteno/uso terapêutico
10.
Blood Adv ; 3(2): 198-211, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670536

RESUMO

It is now recognized that compounds released from tumor cells can activate platelets, causing the release of platelet-derived factors into the tumor microenvironment. Several of these factors have been shown to directly promote neovascularization and metastasis, yet how the feedback between platelet releasate and the tumor cell affects metastatic phenotype remains largely unstudied. Here, we identify that breast tumor cells secrete high levels of interleukin 8 (IL-8, CXCL8) in response to platelet releasate, which promotes their invasive capacity. Furthermore, we found that platelets activate the Akt pathway in breast tumor cells, and inhibition of this pathway eliminated IL-8 production. We therefore hypothesized inhibiting platelets with aspirin could reverse the prometastatic effects of platelets on tumor cell signaling. Platelets treated with aspirin did not activate the Akt pathway, resulting in reduced IL-8 secretion and impaired tumor cell invasion. Of note, patients with breast cancer receiving aspirin had lower circulating IL-8, and their platelets did not increase tumor cell invasion compared with patients not receiving aspirin. Our data suggest platelets support breast tumor metastasis by inducing tumor cells to secrete IL-8. Our data further support that aspirin acts as an anticancer agent by disrupting the communication between platelets and breast tumor cells.


Assuntos
Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Inibidores da Agregação Plaquetária/farmacologia , Plaquetas/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Humanos , Metástase Neoplásica , Estadiamento de Neoplasias , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/farmacologia , Transdução de Sinais
11.
Clin Cancer Res ; 24(10): 2430-2439, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29440174

RESUMO

Purpose: Tumor cell-platelet interactions contribute to tumor progression and metastasis in solid tumors. However, the role of platelets in hematological malignancies is not clear. We investigated the association of platelet activation status with clinical stages in multiple myeloma (MM) patients and explored the role of platelets in MM progression.Experimental Design: Platelets were obtained from healthy donors and MM patients. We examined platelet activation status in MM patients by flow cytometry and transmission electron microscopy. We also observed the enriched pathways that are involved with platelet activation in RNA sequencing of platelets. MM cell lines were used to assess the effect of platelets on MM cell proliferation in vitro and their engraftment in vivo RNA sequencing of MM cell lines was performed to explore molecular mechanisms underlying MM cell-platelet interaction and a CRISPR/Cas9 knockout approach was used for validation.Results: Platelets from MM patients were highly activated with disease progression. RNA sequencing of platelets revealed that genes involved in platelets were enriched in patients with smoldering MM (SMM) or MM. Platelets promoted MM cell proliferation in vitro and contributed to tumor engraftment in bone marrow in vivo RNA sequencing revealed that IL-1ß was upregulated in MM cell lines co-cultured with platelets, whereas IL-1ß knockout in MM cell lines abrogated the effects of platelets on MM cell proliferation and engraftment in vivoConclusions: Platelets from MM patients were highly activated with disease progression. IL-1ß is critical to platelet-mediated MM progression and might be a potential target for MM treatment. Clin Cancer Res; 24(10); 2430-9. ©2018 AACR.


Assuntos
Plaquetas/metabolismo , Regulação Neoplásica da Expressão Gênica , Interleucina-1beta/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Animais , Plaquetas/ultraestrutura , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Interleucina-1beta/metabolismo , Camundongos , Camundongos Transgênicos , Gamopatia Monoclonal de Significância Indeterminada/genética , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/sangue , Mieloma Múltiplo/mortalidade , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Prognóstico
12.
Arterioscler Thromb Vasc Biol ; 37(4): 664-674, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28153880

RESUMO

OBJECTIVE: Platelets, which are mainly known for their role in hemostasis, are now known to play a crucial role in metastasis. Tamoxifen is a selective estrogen receptor modulator that is widely used for the treatment of breast cancer. Tamoxifen and its metabolites have been shown to directly impact platelet function, suggesting that this drug has additional mechanisms of action. The purpose of this study was to determine whether tamoxifen exerts antitumor effects through direct platelet inhibition. APPROACH AND RESULTS: This study found that pretreatment with tamoxifen leads to a significant inhibition of platelet activation. Platelets exposed to tamoxifen released significantly lower amounts of proangiogenic regulator vascular endothelial growth factor. In vitro angiogenesis assays confirmed that tamoxifen pretreatment led to diminished capillary tube formation and decreased endothelial migration. Tamoxifen and its metabolite, 4-hydroxytamoxifen, also significantly inhibited the ability of platelets to promote metastasis in vitro. Using a membrane-based array, we identified several proteins associated with angiogenesis metastasis that were lower in activated releasate from tamoxifen-treated platelets, including angiogenin, chemokine (C-X-C motif) ligand 1, chemokine (C-C motif) ligand 5, epidermal growth factor, chemokine (C-X-C motif) ligand 5, platelet-derived growth factor dimeric isoform BB, whereas antiangiogenic angiopoietin-1 was elevated. Platelets isolated from patients on tamoxifen maintenance therapy were also found to have decreased activation responses, diminished vascular endothelial growth factor release, and lower angiogenic and metastatic potential. CONCLUSIONS: We demonstrate that tamoxifen and its metabolite 4-hydroxytamoxifen directly alter platelet function leading to decreased angiogenic and metastatic potential. Furthermore, this study supports the idea of utilizing targeted platelet therapies to inhibit the platelet's role in angiogenesis and malignancy.


Assuntos
Inibidores da Angiogênese/farmacologia , Plaquetas/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tamoxifeno/análogos & derivados , Plaquetas/metabolismo , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células MCF-7 , Metástase Neoplásica , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Prim Care ; 43(4): 619-635, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27866581

RESUMO

Patients with derangements of secondary hemostasis resulting from inherited or acquired thrombophilias are at increased risk of venous thromboemboli (VTE). Evaluation of a patient with suspected VTE proceeds via evidence-based algorithms that involve computing a pretest probability based on the history and physical examination; this guides subsequent work-up, which can include D dimer and/or imaging. Testing for hypercoagulable disorders should be pursued only in patients with VTE with an increased risk for an underlying thrombophilia. Direct oral anticoagulants are first-line VTE therapies, but they should be avoided in patients who are pregnant, have active cancer, antiphospholipid antibody syndrome, severe renal insufficiency, or prosthetic heart valves.


Assuntos
Anticoagulantes/uso terapêutico , Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/tratamento farmacológico , Atenção Primária à Saúde , Trombose/etiologia , Trombose/prevenção & controle , Contraindicações , Diagnóstico por Imagem , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Testes Hematológicos , Humanos , Medição de Risco , Fatores de Risco
15.
Blood ; 123(1): 101-12, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24065244

RESUMO

Platelets are a reservoir for angiogenic proteins that are secreted in a differentially regulated process. Because of the propensity for clotting, patients with malignancy are often anticoagulated with heparin products, which paradoxically offer a survival benefit by an unknown mechanism. We hypothesized that antithrombotic agents alter the release of angiogenesis regulatory proteins from platelets. Our data revealed that platelets exposed to heparins released significantly decreased vascular endothelial growth factor (VEGF) in response to adenosine 5'-diphosphate or tumor cells (MCF-7 cells) and exhibited a decreased angiogenic potential. The releasate from these platelets contained decreased proangiogenic proteins. The novel anticoagulant fondaparinux (Xa inhibitor) demonstrated a similar impact on the platelet angiogenic potential. Because these anticoagulants decrease thrombin generation, we hypothesized that they disrupt signaling through the platelet protease-activated receptor 1 (PAR1) receptor. Addition of PAR1 antagonists to platelets decreased VEGF release and angiogenic potential. Exposure to a PAR1 agonist in the presence of anticoagulants rescued the angiogenic potential. In vivo studies demonstrated that platelets from anticoagulated patients had decreased VEGF release and angiogenic potential. Our data suggest that the mechanism by which antithrombotic agents increase survival and decrease metastasis in cancer patients is through attenuation of platelet angiogenic potential.


Assuntos
Anticoagulantes/uso terapêutico , Plaquetas/citologia , Neovascularização Patológica/metabolismo , Proteínas Angiogênicas/metabolismo , Anticoagulantes/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Fondaparinux , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , Microscopia de Fluorescência , Inibidores da Agregação Plaquetária/metabolismo , Polissacarídeos/farmacologia , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Cancer Discov ; 2(12): 1150-65, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22896036

RESUMO

UNLABELLED: Breast cancer recurrence rates vary following treatment, suggesting that tumor cells disseminate early from primary sites but remain indolent indefinitely before progressing to symptomatic disease. The reasons why some indolent disseminated tumors erupt into overt disease are unknown. We discovered a novel process by which certain luminal breast cancer (LBC) cells and patient tumor specimens (LBC "instigators") establish a systemic macroenvironment that supports outgrowth of otherwise-indolent disseminated tumors ("responders"). Instigating LBCs secrete cytokines that are absorbed by platelets, which are recruited to responding tumor sites where they aid vessel formation. Instigator-activated bone marrow cells enrich responding tumor cell expression of CD24, an adhesion molecule for platelets, and provide a source of VEGF receptor 2(+) tumor vessel cells. This cascade results in growth of responder adenocarcinomas and is abolished when platelet activation is inhibited by aspirin. These findings highlight the macroenvironment as an important component of disease progression that can be exploited therapeutically. SIGNIFICANCE: Currently, processes that mediate progression of otherwise indolent tumors are not well understood, making it difficult to accurately predict which cancer patients are likely to relapse. Our findings highlight the macroenvironment as an important component of disease progression that can be exploited to more accurately identify patients who would benefit from adjuvant therapy.


Assuntos
Plaquetas/patologia , Células da Medula Óssea/patologia , Neoplasias da Mama/patologia , Animais , Plaquetas/metabolismo , Células da Medula Óssea/metabolismo , Neoplasias da Mama/sangue , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Antígeno CD24/metabolismo , Comunicação Celular/fisiologia , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/sangue , Neovascularização Patológica/patologia , Prognóstico , Transplante Heterólogo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Hematol Oncol Clin North Am ; 26(2): 345-67, ix, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22463831

RESUMO

This article gives a general overview of venous thromboembolism (VTE). Pathophysiology, presentation, diagnosis, and initial management of VTE are briefly reviewed. More difficult management problems are reviewed in greater depth, including duration of anticoagulation, treatment of superficial venous thrombosis, and controversies surrounding bridging therapy, with a brief review of currently available new oral anticoagulants.


Assuntos
Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/terapia , Humanos , Tromboembolia Venosa/etiologia
18.
Blood ; 118(5): 1359-69, 2011 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-21680800

RESUMO

An association between platelets, angiogenesis, and cancer has long been recognized, but the mechanisms linking them remains unclear. Platelets regulate new blood vessel growth through numerous stimulators and inhibitors of angiogenesis by several pathways, including differential exocytosis of angiogenesis regulators. Herein, we investigated the differential release of angiogenesis stimulators and inhibitors from platelets. Activation of human platelets with adenosine diphosphate (ADP) stimulated the release of VEGF, but not endostatin whereas, thromboxane A(2) (TXA(2)) released endostatin but not VEGF. Platelet releasates generated by activation with ADP promoted migration and formation of capillary structures by human umbilical vein endothelial cells (HUV-EC-Cs) in in vitro angiogenesis models. Conversely, TXA(2)-stimulated platelet releasate inhibited migration and formation of capillary structures. Because tumor growth beyond 1-2 mm(3) is angiogenesis-dependent, we hypothesized that cancer cells preferentially stimulate platelets to secrete their pro-angiogenic payload. In support of this, the breast cancer cell line MCF-7 stimulated secretion of VEGF and a pro-angiogenic releasate from platelets. Furthermore, the antiplatelet agent aspirin inhibited platelet-mediated angiogenesis after exposure to ADP or MCF-7 cells providing a potential mechanism for how aspirin may impact malignancy. Manipulation of differentially mediated release of angiogenic factors from platelets may provide a new modality for cancer treatment.


Assuntos
Moduladores da Angiogênese/metabolismo , Moduladores da Angiogênese/farmacologia , Plaquetas/metabolismo , Neovascularização Patológica , Neovascularização Fisiológica/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Difosfato de Adenosina/farmacologia , Aspirina/farmacologia , Neoplasias da Mama/patologia , Carcinoma/patologia , Células Cultivadas , Endostatinas/metabolismo , Endostatinas/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Feminino , Humanos , Modelos Biológicos , Neovascularização Patológica/etiologia , Neovascularização Patológica/prevenção & controle , Vesículas Secretórias/efeitos dos fármacos , Tromboxano A2/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
19.
Curr Hematol Rep ; 4(5): 378-84, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16131439

RESUMO

The risk of venous thromboembolic events (VTE) in patients with cancer is well established. Malignancy screening in patients who present with their first episode of VTE is recommended only if the history or physical findings are suggestive of an underlying problem, however. Thrombotic events remain a significant cause of death in cancer patients and their treatment remains a major challenge in the management of cancer. Low-molecular-weight heparins are safe, effective options for treatment and prophylaxis and may prolong survival in this patient population. It remains to be seen, however, if this treatment will influence cancer outcomes.


Assuntos
Neoplasias/complicações , Trombose Venosa/complicações , Causas de Morte , Humanos , Prognóstico , Fatores de Risco , Resultado do Tratamento , Trombose Venosa/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA