Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 583(7814): 127-132, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32555459

RESUMO

Cellular senescence is characterized by stable cell-cycle arrest and a secretory program that modulates the tissue microenvironment1,2. Physiologically, senescence serves as a tumour-suppressive mechanism that prevents the expansion of premalignant cells3,4 and has a beneficial role in wound-healing responses5,6. Pathologically, the aberrant accumulation of senescent cells generates an inflammatory milieu that leads to chronic tissue damage and contributes to diseases such as liver and lung fibrosis, atherosclerosis, diabetes and osteoarthritis1,7. Accordingly, eliminating senescent cells from damaged tissues in mice ameliorates the symptoms of these pathologies and even promotes longevity1,2,8-10. Here we test the therapeutic concept that chimeric antigen receptor (CAR) T cells that target senescent cells can be effective senolytic agents. We identify the urokinase-type plasminogen activator receptor (uPAR)11 as a cell-surface protein that is broadly induced during senescence and show that uPAR-specific CAR T cells efficiently ablate senescent cells in vitro and in vivo. CAR T cells that target uPAR extend the survival of mice with lung adenocarcinoma that are treated with a senescence-inducing combination of drugs, and restore tissue homeostasis in mice in which liver fibrosis is induced chemically or by diet. These results establish the therapeutic potential of senolytic CAR T cells for senescence-associated diseases.


Assuntos
Envelhecimento/patologia , Senescência Celular/imunologia , Cirrose Hepática/terapia , Longevidade/imunologia , Neoplasias Pulmonares/terapia , Receptores de Antígenos Quiméricos/imunologia , Rejuvenescimento , Linfócitos T/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Animais , Tetracloreto de Carbono , Feminino , Xenoenxertos , Humanos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
3.
Semin Immunol ; 45: 101338, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31744753

RESUMO

In the past several years, a number of C1q binding surface proteins or receptors have been described. This is not of course surprising considering the complexity of the C1q molecule and its ability to bind to a wide range of cellular and plasma proteins via both its collagen-like [cC1q] region and its heterotrimeric globular heads [gC1q] each of which in turn is capable of binding a specific ligand. However, while each of these "receptor" molecules undoubtedly plays a specific function within its restricted microenvironment, and therefore merits full attention, this review nonetheless, will singularly focus on the structure and function of gC1qR-a multi-functional and multi-compartmental protein, which plays an important role in inflammation, infection, and cancer. Although first identified as a receptor for C1q, gC1qR has been shown to bind to a plethora of proteins found in plasma, on the cell surface and on pathogenic microorganisms. The plasma proteins that bind to gC1qR are mostly blood coagulation proteins and include high molecular weight kininogen [HK], Factor XII [Hageman factor], fibrinogen, thrombin [FII], and multimeric vitronectin. This suggests that gC1qR can play an important role in modulating not only of fibrin formation, particularly at local sites of immune injury and/or inflammation, but by activating the kinin/kallikrein system, it is also able to generate, bradykinin, a powerful vasoactive peptide that is largely responsible for the swelling seen in angioedema. Another important function of gC1qR is in cancer, where it has been shown to play a role in tumor cell survival, growth and metastatic invasion by interacting with critical molecules in the tumor cell microenvironment including those of the complement system and kinin system. Finally, by virtue of its ability to interact with a growing list of pathogen-associated molecules, including bacterial and viral ligands, gC1qR is becoming recognized as an important pathogen recognition receptor [PRR]. Given the numerous roles it plays in a growing list of disease settings, gC1qR has now become a potential target for the development of monoclonal antibody-based and/or small molecule-based therapies.


Assuntos
Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Mapeamento Cromossômico , Suscetibilidade a Doenças , Interações Hospedeiro-Parasita , Interações Hospedeiro-Patógeno , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Estrutura Molecular , Receptores de Complemento/química , Receptores de Complemento/genética , Relação Estrutura-Atividade
4.
Ann Neurol ; 90(1): 159-169, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34029423

RESUMO

OBJECTIVE: The objective of this study was to examine the pathophysiology of ischemic stroke with cancer. METHODS: We conducted a prospective cross-sectional study from 2016 to 2020 at 2 hospitals. We enrolled 3 groups of 50 adult participants each. The main group included patients with active solid tumor cancer and acute ischemic stroke. The control groups included patients with acute ischemic stroke only or active cancer only. The patients with stroke-only and patients with cancer-only were matched to the patients with cancer-plus-stroke by age, sex, and cancer type, if applicable. The outcomes were prespecified hematological biomarkers and transcranial Doppler microemboli detection. Hematological biomarkers included markers of coagulation (D-dimer and thrombin-antithrombin), platelet function (P-selectin), and endothelial integrity (thrombomodulin, soluble intercellular adhesion molecule-1 [sICAM-1], and soluble vascular cell adhesion molecule-1 [sVCAM-1]). Hematological biomarkers were compared between groups using the Kruskal-Wallis and Wilcoxon Rank-Sum tests. In multivariable linear regression models, we adjusted for race, number of stroke risk factors, smoking, stroke severity, and antithrombotic use. Transcranial Doppler microemboli presence was compared between groups using chi-square tests. RESULTS: Levels of all study biomarkers were different between groups. In univariate between-group comparisons, patients with cancer-plus-stroke had higher levels of D-dimer, sICAM-1, sVCAM-1, and thrombomodulin than both control groups; higher levels of thrombin-antithrombin than patients with cancer-only; and higher levels of P-selectin than patients with stroke-only. Findings were similar in multivariable analyses. Transcranial Doppler microemboli were detected in 32% of patients with cancer-plus-stroke, 16% of patients with stroke-only, and 6% of patients with cancer-only (p = 0.005). INTERPRETATION: Patients with cancer-related stroke have higher markers of coagulation, platelet, and endothelial dysfunction, and more circulating microemboli, than matched controls. ANN NEUROL 2021;90:159-169.


Assuntos
Encéfalo/diagnóstico por imagem , AVC Isquêmico/complicações , Neoplasias/complicações , Idoso , Biomarcadores/sangue , Estudos Transversais , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio , Humanos , Molécula 1 de Adesão Intercelular/sangue , AVC Isquêmico/sangue , AVC Isquêmico/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/diagnóstico por imagem , Estudos Prospectivos , Trombomodulina/sangue , Ultrassonografia Doppler Transcraniana , Molécula 1 de Adesão de Célula Vascular/sangue
5.
Immunol Rev ; 274(1): 281-289, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27782339

RESUMO

The blood plasma contains four biologically important proteolytic cascades, which probably evolved from the same ancestral gene. This in part may explain why each cascade has very similar "initiating trigger" followed by sequential and cascade-like downstream enzymatic activation pattern. The four cascades are: the complement system, the blood clotting cascade, the fibrinolytic system, and the kallikrein-kinin system. Although much has been written about the interplay between all these enzymatic cascades, the cross-talk between the complement and the kinin generating systems has become particularly relevant as this interaction results in the generation of nascent molecules that have significant impact in various inflammatory diseases including angioedema and cancer. In this review, we will focus on the consequences of the interplay between the two systems by highlighting the role of a novel molecular link called gC1qR. Although this protein was first identified as a receptor for C1q, it is now recognized as a multiligand binding cellular protein, which serves not only as C1q receptor, but also as high affinity (KD  ≤ 0.8 nM) binding site for both high molecular weight kininogen (HK) and factor XII (FXII). At inflammatory sites, where atherogenic factors such as immune complexes and/or pathogens can activate the endothelial cell into a procoagulant and proinflammatory surface, the two pathways are activated to generate vasoactive peptides that contribute in various ways to the inflammatory processes associated with numerous diseases. More importantly, since recent observations strongly suggest an important role for both pathways in cancer, we will focus on how a growing tumor cluster can employ the byproducts derived from the two activation systems to ensure not only its survival and growth, but also its escape into distal sites of colonization.


Assuntos
Angioedema/imunologia , Aterosclerose/imunologia , Complemento C1q/metabolismo , Fator XIa/metabolismo , Imunidade Inata , Inflamação/imunologia , Neoplasias/imunologia , Animais , Humanos , Proteólise , Receptor Cross-Talk , Transdução de Sinais
7.
J Immunol ; 192(1): 377-84, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24319267

RESUMO

Bradykinin (BK) is one of the most potent vasodilator agonists known and belongs to the kinin family of proinflammatory peptides. BK induces its activity via two G protein-coupled receptors: BK receptor 1 (B1R) and BK receptor 2. Although BK receptor 2 is constitutively expressed on endothelial cells (ECs), B1R is induced by IL-1ß. The C1q receptor, receptor for the globular heads of C1q (gC1qR), which plays a role in BK generation, is expressed on activated ECs and is also secreted as soluble gC1qR (sgC1qR). Because sgC1qR can bind to ECs, we hypothesized that it may also serve as an autocrine/paracrine signal for the induction of B1R expression. In this study, we show that gC1qR binds to ECs via a highly conserved domain consisting of residues 174-180, as assessed by solid-phase binding assay and deconvolution fluorescence microscopy. Incubation of ECs (24 h, 37 °C) with sgC1qR resulted in enhancement of B1R expression, whereas incubation with gC1qR lacking aa 174-180 and 154-162 had a diminished effect. Binding of sgC1qR to ECs was through surface-bound fibrinogen and was inhibited by anti-fibrinogen. In summary, our data suggest that, at sites of inflammation, sgC1qR can enhance vascular permeability by upregulation of B1R expression through de novo synthesis, as well as rapid translocation of preformed B1R.


Assuntos
Comunicação Autócrina , Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Proteínas Mitocondriais/metabolismo , Receptor B1 da Bradicinina/metabolismo , Transdução de Sinais , Comunicação Autócrina/efeitos dos fármacos , Sítios de Ligação , Proteínas de Transporte/química , Proteínas de Transporte/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Células Endoteliais/efeitos dos fármacos , Fibrinogênio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Receptor B1 da Bradicinina/genética , Transdução de Sinais/efeitos dos fármacos
8.
Neurocrit Care ; 24(1): 6-46, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26714677

RESUMO

BACKGROUND: The use of antithrombotic agents, including anticoagulants, antiplatelet agents, and thrombolytics has increased over the last decade and is expected to continue to rise. Although antithrombotic-associated intracranial hemorrhage can be devastating, rapid reversal of coagulopathy may help limit hematoma expansion and improve outcomes. METHODS: The Neurocritical Care Society, in conjunction with the Society of Critical Care Medicine, organized an international, multi-institutional committee with expertise in neurocritical care, neurology, neurosurgery, stroke, hematology, hemato-pathology, emergency medicine, pharmacy, nursing, and guideline development to evaluate the literature and develop an evidence-based practice guideline. Formalized literature searches were conducted, and studies meeting the criteria established by the committee were evaluated. RESULTS: Utilizing the GRADE methodology, the committee developed recommendations for reversal of vitamin K antagonists, direct factor Xa antagonists, direct thrombin inhibitors, unfractionated heparin, low-molecular weight heparin, heparinoids, pentasaccharides, thrombolytics, and antiplatelet agents in the setting of intracranial hemorrhage. CONCLUSIONS: This guideline provides timely, evidence-based reversal strategies to assist practitioners in the care of patients with antithrombotic-associated intracranial hemorrhage.


Assuntos
Cuidados Críticos/normas , Fibrinolíticos/administração & dosagem , Fibrinolíticos/efeitos adversos , Hemorragias Intracranianas/induzido quimicamente , Hemorragias Intracranianas/tratamento farmacológico , Neurologia/normas , Guias de Prática Clínica como Assunto/normas , Sociedades Médicas/normas , Humanos
9.
Transfusion ; 55(8): 2001-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25808236

RESUMO

BACKGROUND: Successful peripheral blood stem cell transplantation (PBSCT) depends on the collection and infusion of adequate numbers of peripheral blood progenitor cells (PBPCs). Several predictors of PBPC yield are used currently, including white blood cell (WBC) count and CD34 analysis. This study evaluated the utility of the new automated hematopoietic progenitor cell count available on Sysmex XN hematology analyzers (XN-HPCs) in PBSCT. STUDY DESIGN AND METHODS: The performance characteristics of XN-HPC, CD34+, and WBC analysis were compared using 107 matched peripheral blood and apheresis samples. RESULTS: Good correlation was observed between XN-HPC and CD34+ cell counts in peripheral blood (r = 0.88; slope, 0.81) and apheresis collections (r = 0.91; slope, 0.89). Moreover, peripheral blood XN-HPC and CD34 analysis showed comparable ability to predict successful PBPC harvests (≥2 × 10(6) CD34+ cells/kg). At a cutoff of 20 × 10(6) progenitor cells/L, peripheral blood XN- HPC and CD34 analysis both showed negative predictive values (NPVs) of 100% and positive predictive values (PPVs) of 55.4 and 63%, respectively. Using an optimized cutoff of 38 × 10(6) progenitor cells/L, derived from receiver operating characteristic analysis, the PPV for XN-HPC and CD34 analysis increased to 71.4 and 78.9%, respectively, with relatively unchanged NPVs (XN-HPC 97.7%, CD34+ 98.0%). In contrast, the correlation between peripheral blood WBC and CD34 analysis was poor (r = 0.48; slope, 669.85), and the peripheral blood WBC count (cutoff, 10 × 10(9) /L) was a poor predictor of PBPC harvest (NPV 60%, PPV 43.1%). CONCLUSION: XN-HPC compares favorably with CD34 analysis and may be a surrogate for CD34 analysis to predict optimal timing of PBPC collections.


Assuntos
Contagem de Células Sanguíneas/instrumentação , Células-Tronco Hematopoéticas/citologia , Transplante de Células-Tronco de Sangue Periférico , Adolescente , Adulto , Idoso , Antígenos CD34/sangue , Automação , Remoção de Componentes Sanguíneos , Núcleo Celular/ultraestrutura , Tamanho Celular , Criança , Pré-Escolar , Grânulos Citoplasmáticos/ultraestrutura , Desenho de Equipamento , Feminino , Citometria de Fluxo/instrumentação , Mobilização de Células-Tronco Hematopoéticas , Humanos , Lactente , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Curva ROC , Reprodutibilidade dos Testes , Adulto Jovem
10.
Blood ; 120(6): 1228-36, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22700724

RESUMO

C1q modulates the differentiation and function of cells committed to the monocyte-derived dendritic cell (DC) lineage. Because the 2 C1q receptors found on the DC surface-gC1qR and cC1qR-lack a direct conduit into intracellular elements, we postulated that the receptors must form complexes with transmembrane partners. In the present study, we show that DC-SIGN, a C-type lectin expressed on DCs, binds directly to C1q, as assessed by ELISA, flow cytometry, and immunoprecipitation experiments. Surface plasmon resonance analysis revealed that the interaction was specific, and both intact C1q and the globular portion of C1q bound to DC-SIGN. Whereas IgG reduced this binding significantly, the Arg residues (162-163) of the C1q-A chain, which are thought to contribute to the C1q-IgG interaction, were not required for C1q binding to DC-SIGN. Binding was reduced significantly in the absence of Ca(2+) and by preincubation of DC-SIGN with mannan, suggesting that C1q binds to DC-SIGN at its principal Ca(2+)-binding pocket, which has increased affinity for mannose residues. Antigen-capture ELISA and immunofluorescence microscopy revealed that C1q and gC1qR associate with DC-SIGN on blood DC precursors and immature DCs. The results of the present study suggest that C1q/gC1qR may regulate DC differentiation and function through the DC-SIGN-mediated induction of cell-signaling pathways.


Assuntos
Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/metabolismo , Complemento C1q/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Proteínas Mitocondriais/metabolismo , Complexos Multiproteicos/metabolismo , Receptores de Superfície Celular/metabolismo , Ligação Competitiva , Cálcio/farmacologia , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/fisiologia , Humanos , Modelos Biológicos , Monócitos/metabolismo , Monócitos/fisiologia , Complexos Multiproteicos/fisiologia , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/fisiologia
11.
Front Immunol ; 15: 1351656, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38711524

RESUMO

Understanding at the molecular level of the cell biology of tumors has led to significant treatment advances in the past. Despite such advances however, development of therapy resistance and tumor recurrence are still unresolved major challenges. This therefore underscores the need to identify novel tumor targets and develop corresponding therapies to supplement existing biologic and cytotoxic approaches so that a deeper and more sustained treatment responses could be achieved. The complement system is emerging as a potential novel target for cancer therapy. Data accumulated to date show that complement proteins, and in particular C1q and its receptors cC1qR/CR and gC1qR/p33/HABP1, are overexpressed in most cancer cells and together are involved not only in shaping the inflammatory tumor microenvironment, but also in the regulation of angiogenesis, metastasis, and cell proliferation. In addition to the soluble form of C1q that is found in plasma, the C1q molecule is also found anchored on the cell membrane of monocytes, macrophages, dendritic cells, and cancer cells, via a 22aa long leader peptide found only in the A-chain. This orientation leaves its 6 globular heads exposed outwardly and thus available for high affinity binding to a wide range of molecular ligands that enhance tumor cell survival, migration, and proliferation. Similarly, the gC1qR molecule is not only overexpressed in most cancer types but is also released into the microenvironment where it has been shown to be associated with cancer cell proliferation and metastasis by activation of the complement and kinin systems. Co-culture of either T cells or cancer cells with purified C1q or anti-gC1qR has been shown to induce an anti-proliferative response. It is therefore postulated that in the tumor microenvironment, the interaction between C1q expressing cancer cells and gC1qR bearing cytotoxic T cells results in T cell suppression in a manner akin to the PD-L1 and PD-1 interaction.


Assuntos
Proteínas de Transporte , Complemento C1q , Inibidores de Checkpoint Imunológico , Glicoproteínas de Membrana , Proteínas Mitocondriais , Neoplasias , Receptores de Complemento , Humanos , Complemento C1q/metabolismo , Complemento C1q/imunologia , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Complemento/metabolismo , Animais , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Microambiente Tumoral/imunologia
12.
Adv Exp Med Biol ; 735: 97-110, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402021

RESUMO

Abstract The receptor for the globular heads of C1q, gC1qR/p33, is a widely expressed cellular protein, which binds to diverse ligands including plasma proteins, cellular proteins, and microbial ligands. In addition to C1q, gC1qR also binds high molecular weight kininogen (HK), which also has two other cell surface sites, namely, cytokeratin 1 and urokinase plasminogen activator receptor (uPAR). On endothelial cells (ECs), the three molecules form two closely associated bimolecular complexes of gC1qR/cytokeratin 1 and uPAR/cytokeratin 1. However, by virtue of its high affinity for HK, gC1qR plays a central role in the assembly of the kallikrein-kinin system, leading to the generation of bradykinin (BK). BK in turn is largely responsible for the vascular leakage and associated inflammation seen in angioedema patients. Therefore, blockade of gC1qR by inhibitory peptides or antibodies may not only prevent the generation of BK but also reduce Clq-induced or microbial-ligand-induced inflammatory responses. Employing synthetic peptides and gClqR deletion mutants, we confirmed previously predicted sites for C1q (residues 75-96) and HK (residues 204-218) and identified additional sites for both C1q and HK (residues 190-202), for C1q (residues 144-162), and for HIV-1 gp41 (residues 174-180). With the exception of residues 75-96, which is located in the alphaA coiled-coil N-terminal segment, most of the identified residues form part of the highly charged loops connecting the various beta-strands in the crystal structure. Taken together, the data support the notion that gC1qR could serve as a novel molecular target for the design of antibody-based and/or peptide-based therapy to attenuate acute and/or chronic inflammation associated with vascular leakage and infection.


Assuntos
Complemento C1q/efeitos dos fármacos , Infecções/tratamento farmacológico , Inflamação/tratamento farmacológico , Receptores de Complemento/efeitos dos fármacos , Animais , Humanos , Ligantes , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Receptores de Complemento/química , Receptores de Complemento/genética
13.
J Mol Diagn ; 24(8): 867-877, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35934321

RESUMO

Detection of serum embryonic miRNAs miR-371a-3p and miR-372-3p has been proposed to aid in diagnosis, prognosis, and management of patients with testicular germ cell tumors (GCTs). This study describes the analytical validation and performance of a laboratory-developed test to detect these miRNA targets by stem loop real-time quantitative RT-PCR (RT-qPCR) in serum from patients with GCTs. The assay was standardized using an exogenous spike-in control of nonhuman miRNA from Caenorhabditis elegans (cel-miR-39-3p) to assess extraction efficiency, and an endogenous housekeeping miRNA, miR-30b-5p, to control for miRNA normalization. miRNA results were expressed as relative expression level, using the comparative threshold cycle method (2ΔΔCT). Analytical sensitivity of miR-371a-3p and miR-372-3p was 12.5 and 1.25 copies/µL, respectively. Clinical accuracy was evaluated using GCT patients with (n = 34) and without (n = 17) active disease. Positive/negative cutoffs and indeterminate findings were established on the basis of results from healthy volunteers (n = 25) and assay precision. miR-371a-3p and miR-372-3p exhibited a sensitivity of 81.8% and 87.5%, respectively, and a specificity of 100% and 94%, respectively, and an area under the receiver operating characteristic curve of 0.93 and 0.95, respectively. Taken together, RT-qPCR testing for serum miR-371a-3p and miR-372-3p represents a robust, sensitive, and specific clinical assay to aid in the clinical management of patients with GCT.


Assuntos
MicroRNAs , Neoplasias Embrionárias de Células Germinativas , Biomarcadores Tumorais/genética , Humanos , Laboratórios Clínicos , Masculino , Neoplasias Embrionárias de Células Germinativas/diagnóstico , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Testiculares
14.
J Thromb Haemost ; 20(9): 2046-2057, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35652416

RESUMO

BACKGROUND: Patients with cancer and acute ischemic stroke (AIS) face high rates of recurrent thromboembolism or death. OBJECTIVES: To examine whether hematologic and embolic biomarkers soon after AIS are associated with subsequent adverse clinical outcomes. METHODS: We prospectively enrolled 50 adults with active solid tumor cancer and AIS at two hospitals from 2016 to 2020. Blood was collected 72-120 h after stroke onset. A 30-min transcranial Doppler (TCD) microemboli detection study was performed. The exposure variables were hematologic markers of coagulation (D-dimer, thrombin-antithrombin), platelet (P-selectin), and endothelial activation (thrombomodulin, soluble intercellular adhesion molecule-1 [sICAM-1], soluble vascular cell adhesion molecule-1 [sVCAM-1]), and the presence of TCD microemboli. The primary outcome was a composite of recurrent arterial/venous thromboembolism or death. We used Cox regression to evaluate associations between biomarkers and subsequent outcomes. RESULTS: During an estimated median follow-up time of 48 days (IQR, 18-312), 43 (86%) participants developed recurrent thromboembolism or death, including 28 (56%) with recurrent thromboembolism, of which 13 were recurrent AIS (26%). In unadjusted analysis, D-dimer (HR 1.6; 95% CI 1.2-2.0), P-selectin (HR 1.9; 95% CI 1.4-2.7), sICAM-1 (HR 2.2; 95% CI 1.6-3.1), sVCAM-1 (HR 1.6; 95% CI 1.2-2.1), and microemboli (HR 2.2; 95% CI 1.1-4.5) were associated with the primary outcome, whereas thrombin-antithrombin and thrombomodulin were not. D-dimer was the only marker associated with recurrent AIS (HR 1.2; 95% CI 1.0-1.5). Results were generally consistent in analyses adjusted for important prognostic variables. CONCLUSIONS: Markers of hypercoagulability and embolic disease may be associated with adverse clinical outcomes in cancer-related stroke.


Assuntos
AVC Isquêmico , Neoplasias , Tromboembolia , Adulto , Antitrombinas , Biomarcadores , Humanos , AVC Isquêmico/diagnóstico , Neoplasias/complicações , Selectina-P , Trombina , Trombomodulina
16.
J Neuroinflammation ; 8: 103, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21854561

RESUMO

BACKGROUND: Subarachnoid haemorrhage (SAH) elicits rapid pathological changes in the structure and function of parenchymal vessels (≤ 100 µm). The role of neutrophils in these changes has not been determined. This study investigates the role of neutrophils in early microvascular changes after SAH METHOD: Rats were either untreated, treated with vinblastine or anti-polymorphonuclear (PMN) serum, which depletes neutrophils, or treated with pyrrolidine dithiocarbamate (PDTC), which limits neutrophil activity. SAH was induced by endovascular perforation. Neutrophil infiltration and the integrity of vascular endothelium and basement membrane were assessed immunohistochemically. Vascular collagenase activity was assessed by in situ zymography. RESULTS: Vinblastine and anti-PMN serum reduced post-SAH accumulation of neutrophils in cerebral vessels and in brain parenchyma. PDTC increased the neutrophil accumulation in cerebral vessels and decreased accumulation in brain parenchyma. In addition, each of the three agents decreased vascular collagenase activity and post-SAH loss of vascular endothelial and basement membrane immunostaining. CONCLUSIONS: Our results implicate neutrophils in early microvascular injury after SAH and indicate that treatments which reduce neutrophil activity can be beneficial in limiting microvascular injury and increasing survival after SAH.


Assuntos
Microcirculação , Ativação de Neutrófilo , Neutrófilos/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Biomarcadores/metabolismo , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Permeabilidade Capilar , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Masculino , Infiltração de Neutrófilos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Prolina/análogos & derivados , Prolina/metabolismo , Prolina/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/fisiopatologia , Tiocarbamatos/metabolismo , Tiocarbamatos/farmacologia , Moduladores de Tubulina/farmacologia , Vimblastina/farmacologia
18.
Microvasc Res ; 82(1): 66-72, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21539847

RESUMO

Endovascular infections with Staphylococcus aureus (S. aureus) are associated with high mortality. gC1qR/p33 (gC1qR), a receptor for the complement component C1q expressed on endothelial cells, interacts with protein A of S. aureus and gC1qR blockade reduces S. aureus colonization during infective endocarditis. The aim of this study was to analyze in vivo whether this observation is due to a decreased interaction of S. aureus with the microvascular endothelium. A dorsal skinfold chamber was prepared in Syrian golden hamsters, which were treated with the monoclonal antibody (MAb) 74.5.2 directed against gC1qR or vehicle. The interaction of fluorescein isothiocyanate (FITC)-labeled staphylococci and leukocytes with the endothelium was analyzed under physiological conditions as well as after TNF-α-induced inflammation using intravital fluorescence microscopy. Administration of MAb 74.5.2 significantly reduced adherence of S. aureus to the endothelium in untreated and TNF-α-exposed tissue. In addition, we could demonstrate in vitro that S. aureus adherence to human endothelial cells was inhibited by MAb 74.5.2. Blockade of gC1qR did not affect leukocyte-endothelial cell interaction. In conclusion, our findings indicate that immunological inhibition of gC1qR may be therapeutically used to decrease the interaction of S. aureus with the microvascular endothelium.


Assuntos
Adesão Celular/fisiologia , Endotélio Vascular/microbiologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Microvasos/microbiologia , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/metabolismo , Staphylococcus aureus/citologia , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Procedimentos Cirúrgicos Dermatológicos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/microbiologia , Endotélio Vascular/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/química , Humanos , Inflamação/induzido quimicamente , Inflamação/microbiologia , Inflamação/patologia , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Leucócitos/patologia , Masculino , Glicoproteínas de Membrana/imunologia , Mesocricetus , Microscopia de Fluorescência , Microvasos/efeitos dos fármacos , Receptores de Complemento/imunologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Pele/irrigação sanguínea , Coloração e Rotulagem , Fator de Necrose Tumoral alfa/farmacologia
19.
J Immunol ; 183(9): 6013-20, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19828637

RESUMO

Mixed cryoglobulinemia (MC) is a lymphoproliferative disorder observed in approximately 10 to 15% of hepatitis C virus (HCV)-infected patients. Circulating, nonenveloped HCV core protein, which has been detected in cryoprecipitable immune complexes, interacts with immunocytes through the receptor for the globular domain of C1q protein (gC1q-R). In this study, we have evaluated circulating gC1q-R levels in chronically HCV-infected patients, with and without MC. These levels were significantly higher in MC patients than in those without MC and in healthy controls and paralleled specific mRNA expression in PBL. Soluble gC1q-R circulates as a complexed form containing both C1q and HCV core proteins. Higher serum gC1q-R levels negatively correlated with circulating concentrations of the C4d fragment. The presence of sequestered C4d in the vascular bed of skin biopsies from MC patients was indicative of in situ complement activation. In vitro studies showed that release of soluble gC1q-R is regulated by HCV core-mediated inhibition of cell proliferation. Our results indicate that up-regulation of gC1q-R expression is a distinctive feature of MC, and that dysregulated shedding of C1q-R molecules contributes to vascular cryoglobulin-induced damage via the classic complement-mediated pathway.


Assuntos
Complemento C1q/metabolismo , Crioglobulinemia/imunologia , Crioglobulinas/efeitos adversos , Hepacivirus/imunologia , Hepatite C Crônica/imunologia , Glicoproteínas de Membrana/fisiologia , Receptores de Complemento/fisiologia , Vasculite/imunologia , Via Clássica do Complemento/imunologia , Crioglobulinemia/metabolismo , Crioglobulinemia/virologia , Feminino , Hepatite C Crônica/metabolismo , Hepatite C Crônica/virologia , Humanos , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/sangue , Pessoa de Meia-Idade , Estrutura Terciária de Proteína , Receptores de Complemento/biossíntese , Receptores de Complemento/sangue , Regulação para Cima/imunologia , Vasculite/metabolismo , Vasculite/virologia , Proteínas do Core Viral/imunologia , Proteínas do Core Viral/metabolismo
20.
Front Immunol ; 12: 716361, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34491250

RESUMO

Background: COVID-19 pathology is associated with exuberant inflammation, vascular damage, and activation of coagulation. In addition, complement activation has been described and is linked to disease pathology. However, few studies have been conducted in cancer patients. Objective: This study examined complement activation in response to COVID-19 in the setting of cancer associated thromboinflammation. Methods: Markers of complement activation (C3a, C5a, sC5b-9) and complement inhibitors (Factor H, C1-Inhibitor) were evaluated in plasma of cancer patients with (n=43) and without (n=43) COVID-19 and stratified based on elevated plasma D-dimer levels (>1.0 µg/ml FEU). Markers of vascular endothelial cell dysfunction and platelet activation (ICAM-1, thrombomodulin, P-selectin) as well as systemic inflammation (pentraxin-3, serum amyloid A, soluble urokinase plasminogen activator receptor) were analyzed to further evaluate the inflammatory response. Results: Increases in circulating markers of endothelial cell dysfunction, platelet activation, and systemic inflammation were noted in cancer patients with COVID-19. In contrast, complement activation increased in cancer patients with COVID-19 and elevated D-dimers. This was accompanied by decreased C1-Inhibitor levels in patients with D-dimers > 5 ug/ml FEU. Conclusion: Complement activation in cancer patients with COVID-19 is significantly increased in the setting of thromboinflammation. These findings support a link between coagulation and complement cascades in the setting of inflammation.


Assuntos
COVID-19/imunologia , Ativação do Complemento/imunologia , Inflamação/imunologia , Neoplasias/imunologia , SARS-CoV-2/imunologia , Trombose/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , COVID-19/sangue , COVID-19/virologia , Inativadores do Complemento/sangue , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Humanos , Inflamação/sangue , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Ativação Plaquetária/imunologia , Estudos Retrospectivos , SARS-CoV-2/fisiologia , Trombose/sangue , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA