Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Nature ; 580(7804): 524-529, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32322056

RESUMEN

The initiation of an intestinal tumour is a probabilistic process that depends on the competition between mutant and normal epithelial stem cells in crypts1. Intestinal stem cells are closely associated with a diverse but poorly characterized network of mesenchymal cell types2,3. However, whether the physiological mesenchymal microenvironment of mutant stem cells affects tumour initiation remains unknown. Here we provide in vivo evidence that the mesenchymal niche controls tumour initiation in trans. By characterizing the heterogeneity of the intestinal mesenchyme using single-cell RNA-sequencing analysis, we identified a population of rare pericryptal Ptgs2-expressing fibroblasts that constitutively process arachidonic acid into highly labile prostaglandin E2 (PGE2). Specific ablation of Ptgs2 in fibroblasts was sufficient to prevent tumour initiation in two different models of sporadic, autochthonous tumorigenesis. Mechanistically, single-cell RNA-sequencing analyses of a mesenchymal niche model showed that fibroblast-derived PGE2 drives the expansion οf a population of Sca-1+ reserve-like stem cells. These express a strong regenerative/tumorigenic program, driven by the Hippo pathway effector Yap. In vivo, Yap is indispensable for Sca-1+ cell expansion and early tumour initiation and displays a nuclear localization in both mouse and human adenomas. Using organoid experiments, we identified a molecular mechanism whereby PGE2 promotes Yap dephosphorylation, nuclear translocation and transcriptional activity by signalling through the receptor Ptger4. Epithelial-specific ablation of Ptger4 misdirected the regenerative reprogramming of stem cells and prevented Sca-1+ cell expansion and sporadic tumour initiation in mutant mice, thereby demonstrating the robust paracrine control of tumour-initiating stem cells by PGE2-Ptger4. Analyses of patient-derived organoids established that PGE2-PTGER4 also regulates stem-cell function in humans. Our study demonstrates that initiation of colorectal cancer is orchestrated by the mesenchymal niche and reveals a mechanism by which rare pericryptal Ptgs2-expressing fibroblasts exert paracrine control over tumour-initiating stem cells via the druggable PGE2-Ptger4-Yap signalling axis.


Asunto(s)
Carcinogénesis , Neoplasias Colorrectales/patología , Intestinos/patología , Mesodermo/patología , Células Madre Neoplásicas/patología , Comunicación Paracrina , Nicho de Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos Ly/metabolismo , Ácido Araquidónico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Proteínas de la Membrana/metabolismo , Mesodermo/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Organoides/metabolismo , Organoides/patología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Análisis de la Célula Individual , Proteínas Señalizadoras YAP
2.
Arterioscler Thromb Vasc Biol ; 44(1): 271-286, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823267

RESUMEN

BACKGROUND: Prostacyclin is a fundamental signaling pathway traditionally associated with the cardiovascular system and protection against thrombosis but which also has regulatory functions in fibrosis, proliferation, and immunity. Prevailing dogma states that prostacyclin is principally derived from vascular endothelium, although it is known that other cells can also synthesize it. However, the role of nonendothelial sources in prostacyclin production has not been systematically evaluated resulting in an underappreciation of their importance relative to better characterized endothelial sources. METHODS: To address this, we have used novel endothelial cell-specific and fibroblast-specific COX (cyclo-oxygenase) and prostacyclin synthase knockout mice and cells freshly isolated from mouse and human lung tissue. We have assessed prostacyclin release by immunoassay and thrombosis in vivo using an FeCl3-induced carotid artery injury model. RESULTS: We found that in arteries, endothelial cells are the main source of prostacyclin but that in the lung, and other tissues, prostacyclin production occurs largely independently of endothelial and vascular smooth muscle cells. Instead, in mouse and human lung, prostacyclin production was strongly associated with fibroblasts. By comparison, microvascular endothelial cells from the lung showed weak prostacyclin synthetic capacity compared with those isolated from large arteries. Prostacyclin derived from fibroblasts and other nonendothelial sources was seen to contribute to antithrombotic protection. CONCLUSIONS: These observations define a new paradigm in prostacyclin biology in which fibroblast/nonendothelial-derived prostacyclin works in parallel with endothelium-derived prostanoids to control thrombotic risk and potentially a broad range of other biology. Although generation of prostacyclin by fibroblasts has been shown previously, the scale and systemic activity was unappreciated. As such, this represents a basic change in our understanding and may provide new insight into how diseases of the lung result in cardiovascular risk.


Asunto(s)
Epoprostenol , Trombosis , Ratones , Humanos , Animales , Fibrinolíticos , Células Endoteliales/metabolismo , Prostaglandinas I/metabolismo , Prostaglandinas I/farmacología , Endotelio Vascular/metabolismo , Ratones Noqueados , Fibroblastos/metabolismo , Trombosis/genética , Trombosis/prevención & control , Trombosis/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836566

RESUMEN

Unlike other epithelial cancer types, circulating tumor cells (CTCs) are less frequently detected in the peripheral blood of non-small cell lung cancer (NSCLC) patients using epithelial marker-based detection approaches despite the aggressive nature of NSCLC. Here, we demonstrate hexokinase-2 (HK2) as a metabolic function-associated marker for the detection of CTCs. In 59 NSCLC patients bearing cytokeratin-positive (CKpos) primary tumors, HK2 enables resolving cytokeratin-negative (HK2high/CKneg) CTCs as a prevalent population in about half of the peripheral blood samples with positive CTC counts. However, HK2high/CKneg tumor cells are a minority population in pleural effusions and cerebrospinal fluids. Single-cell analysis shows that HK2high/CKneg CTCs exhibit smaller sizes but consistent copy number variation profiles compared with CKpos counterparts. Single-cell transcriptome profiling reveals that CK expression levels of CTCs are independent of their epithelial-to-mesenchymal transition (EMT) status, challenging the long-standing association between CK expression and EMT. HK2high/CKneg CTCs display metastasis and EGFR inhibitor resistance-related molecular signatures and are selectively enriched in patients with EGFRL858R driver oncogene mutation as opposed to EGFR19Del , which is more frequently found in patients with prevalent CKpos CTCs in the blood. Consistently, treatment-naïve patients with a larger number or proportion of HK2high/CKneg CTCs in the blood exhibit poor therapy response and shorter progression-free survival. Collectively, our approach resolves a more complete spectrum of CTCs in NSCLC that can potentially be exploited to identify patient prognosis before therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Hexoquinasa/sangre , Neoplasias Pulmonares/patología , Células Neoplásicas Circulantes/patología , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Transición Epitelial-Mesenquimal , Receptores ErbB/genética , Genotipo , Humanos , Queratinas/sangre , Biopsia Líquida , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/enzimología , Pronóstico
4.
FASEB J ; 34(8): 10027-10040, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32592197

RESUMEN

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Asunto(s)
Aspirina/farmacología , Plaquetas/metabolismo , Ciclooxigenasa 1/fisiología , Inhibidores de la Ciclooxigenasa/farmacología , Eicosanoides/metabolismo , Proteínas de la Membrana/fisiología , Trombosis/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , Plaquetas/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombosis/tratamiento farmacológico , Trombosis/patología
5.
Circ Res ; 125(9): 847-854, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31510878

RESUMEN

RATIONALE: Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo. OBJECTIVE: We sought to identify the role, if any, of COX1-derived prostacyclin in antithrombotic protection in vivo and compare this to the established protective role of COX2. METHODS AND RESULTS: We developed vascular-specific COX1 knockout mice and studied them alongside endothelial-specific COX2 knockout mice. COX1 immunoreactivity and prostacyclin production were primarily associated with the endothelial layer of aortae; freshly isolated aortic ECs released >10-fold more prostacyclin than smooth muscle cells. Moreover, aortic prostacyclin production, the ability of aortic rings to inhibit platelet aggregation and plasma prostacyclin levels were reduced when COX1 was knocked out in ECs but not in smooth muscle cells. When thrombosis was measured in vivo after FeCl3 carotid artery injury, endothelial COX1 deletion accelerated thrombosis to a similar extent as prostacyclin receptor blockade. However, this effect was lost when COX1 was deleted from both ECs and platelets. Deletion of COX2 from ECs also resulted in a prothrombotic phenotype that was independent of local vascular prostacyclin production. CONCLUSIONS: These data demonstrate for the first time that, in healthy animals, endothelial COX1 provides an essential antithrombotic tone, which is masked when COX1 activity is lost in both ECs and platelets. These results help us define a new 2-component paradigm wherein thrombotic tone is regulated by both COX1 and COX2 through complementary but mechanistically distinct pathways.


Asunto(s)
Ciclooxigenasa 1/deficiencia , Endotelio Vascular/metabolismo , Epoprostenol/metabolismo , Fibrinolíticos/metabolismo , Eliminación de Gen , Proteínas de la Membrana/deficiencia , Agregación Plaquetaria/fisiología , Animales , Aorta/metabolismo , Ciclooxigenasa 1/genética , Epoprostenol/genética , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos
6.
Proc Natl Acad Sci U S A ; 113(2): 434-9, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26712011

RESUMEN

Cyclooxygenase-2 (COX-2) is an inducible enzyme that drives inflammation and is the therapeutic target for widely used nonsteroidal antiinflammatory drugs (NSAIDs). However, COX-2 is also constitutively expressed, in the absence of overt inflammation, with a specific tissue distribution that includes the kidney, gastrointestinal tract, brain, and thymus. Constitutive COX-2 expression is therapeutically important because NSAIDs cause cardiovascular and renal side effects in otherwise healthy individuals. These side effects are now of major concern globally. However, the pathways driving constitutive COX-2 expression remain poorly understood. Here we show that in the kidney and other sites, constitutive COX-2 expression is a sterile response, independent of commensal microorganisms and not associated with activity of the inflammatory transcription factor NF-κB. Instead, COX-2 expression in the kidney but not other regions colocalized with nuclear factor of activated T cells (NFAT) transcription factor activity and was sensitive to inhibition of calcineurin-dependent NFAT activation. However, calcineurin/NFAT regulation did not contribute to constitutive expression elsewhere or to inflammatory COX-2 induction at any site. These data address the mechanisms driving constitutive COX-2 and suggest that by targeting transcription it may be possible to develop antiinflammatory therapies that spare the constitutive expression necessary for normal homeostatic functions, including those important to the cardiovascular-renal system.


Asunto(s)
Ciclooxigenasa 2/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Transducción de Señal , Transcripción Genética , Animales , Ciclooxigenasa 2/metabolismo , Ciclosporina/farmacología , Citocinas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Vida Libre de Gérmenes , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipopolisacáridos/farmacología , Luciferasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
7.
Proc Natl Acad Sci U S A ; 113(15): 4027-32, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035974

RESUMEN

Deoxycytidine kinase (dCK), a rate-limiting enzyme in the cytosolic deoxyribonucleoside (dN) salvage pathway, is an important therapeutic and positron emission tomography (PET) imaging target in cancer. PET probes for dCK have been developed and are effective in mice but have suboptimal specificity and sensitivity in humans. To identify a more suitable probe for clinical dCK PET imaging, we compared the selectivity of two candidate compounds-[(18)F]Clofarabine; 2-chloro-2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-adenine ([(18)F]CFA) and 2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-guanine ([(18)F]F-AraG)-for dCK and deoxyguanosine kinase (dGK), a dCK-related mitochondrial enzyme. We demonstrate that, in the tracer concentration range used for PET imaging, [(18)F]CFA is primarily a substrate for dCK, with minimal cross-reactivity. In contrast, [(18)F]F-AraG is a better substrate for dGK than for dCK. [(18)F]CFA accumulation in leukemia cells correlated with dCK expression and was abrogated by treatment with a dCK inhibitor. Although [(18)F]CFA uptake was reduced by deoxycytidine (dC) competition, this inhibition required high dC concentrations present in murine, but not human, plasma. Expression of cytidine deaminase, a dC-catabolizing enzyme, in leukemia cells both in cell culture and in mice reduced the competition between dC and [(18)F]CFA, leading to increased dCK-dependent probe accumulation. First-in-human, to our knowledge, [(18)F]CFA PET/CT studies showed probe accumulation in tissues with high dCK expression: e.g., hematopoietic bone marrow and secondary lymphoid organs. The selectivity of [(18)F]CFA for dCK and its favorable biodistribution in humans justify further studies to validate [(18)F]CFA PET as a new cancer biomarker for treatment stratification and monitoring.


Asunto(s)
Nucleótidos de Adenina/química , Arabinonucleósidos/química , Biomarcadores de Tumor/química , Desoxicitidina Quinasa/análisis , Desoxicitidina Quinasa/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Antineoplásicos/química , Línea Celular Tumoral , Clofarabina , Medios de Contraste/química , Desoxicitidina Quinasa/antagonistas & inhibidores , Humanos , Leucemia/enzimología , Ratones , Neoplasias/tratamiento farmacológico , Profármacos/química , Ratas
8.
J Virol ; 90(1): 412-20, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491162

RESUMEN

UNLABELLED: Adeno-associated virus 2 (AAV2) and adenovirus 5 (Ad5) are promising gene therapy vectors. Both display liver tropism and are currently thought to enter hepatocytes in vivo through cell surface heparan sulfate proteoglycans (HSPGs). To test directly this hypothesis, we created mice that lack Ext1, an enzyme required for heparan sulfate biosynthesis, in hepatocytes. Ext1(HEP) mutant mice exhibit an 8-fold reduction of heparan sulfate in primary hepatocytes and a 5-fold reduction of heparan sulfate in whole liver tissue. Conditional hepatocyte Ext1 gene deletion greatly reduced AAV2 liver transduction following intravenous injection. Ad5 transduction requires blood coagulation factor X (FX); FX binds to the Ad5 capsid hexon protein and bridges the virus to HSPGs on the cell surface. Ad5.FX transduction was abrogated in primary hepatocytes from Ext1(HEP) mice. However, in contrast to the case with AAV2, Ad5 transduction was not significantly reduced in the livers of Ext1(HEP) mice. FX remained essential for Ad5 transduction in vivo in Ext1(HEP) mice. We conclude that while AAV2 requires HSPGs for entry into mouse hepatocytes, HSPGs are dispensable for Ad5 hepatocyte transduction in vivo. This study reopens the question of how adenovirus enters cells in vivo. IMPORTANCE: Our understanding of how viruses enter cells, and how they can be used as therapeutic vectors to manage disease, begins with identification of the cell surface receptors to which viruses bind and which mediate viral entry. Both adeno-associated virus 2 and adenovirus 5 are currently thought to enter hepatocytes in vivo through heparan sulfate proteoglycans (HSPGs). However, direct evidence for these conclusions is lacking. Experiments presented herein, in which hepatic heparan sulfate synthesis was genetically abolished, demonstrated that HSPGs are not likely to function as hepatocyte Ad5 receptors in vivo. The data also demonstrate that HSPGs are required for hepatocyte transduction by AAV2. These results reopen the question of the identity of the Ad5 receptor in vivo and emphasize the necessity of demonstrating the nature of the receptor by genetic means, both for understanding Ad5 entry into cells in vivo and for optimization of Ad5 vectors as therapeutic agents.


Asunto(s)
Adenoviridae/genética , Dependovirus/genética , Heparitina Sulfato/metabolismo , Hepatocitos/virología , Hígado/virología , Receptores Virales/metabolismo , Transducción Genética , Animales , Células Cultivadas , Femenino , Vectores Genéticos , Hepatocitos/química , Hígado/química , Masculino , Ratones
9.
Proc Natl Acad Sci U S A ; 111(43): E4658-67, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25316791

RESUMEN

Tumor progression locus-2 (Tpl2) kinase is a major inflammatory mediator in immune cell types recently found to be genetically associated with inflammatory bowel diseases (IBDs). Here we show that Tpl2 may exert a dominant homeostatic rather than inflammatory function in the intestine mediated specifically by subepithelial intestinal myofibroblasts (IMFs). Mice with complete or IMF-specific Tpl2 ablation are highly susceptible to epithelial injury-induced colitis showing impaired compensatory proliferation in crypts and extensive ulcerations without significant changes in inflammatory responses. Following epithelial injury, IMFs sense innate or inflammatory signals and activate, via Tpl2, the cyclooxygenase-2 (Cox-2)-prostaglandin E2 (PGE2) pathway, which we show here to be essential for the epithelial homeostatic response. Exogenous PGE2 administration rescues mice with complete or IMF-specific Tpl2 ablation from defects in crypt function and susceptibility to colitis. We also show that Tpl2 expression is decreased in IMFs isolated from the inflamed ileum of IBD patients indicating that Tpl2 function in IMFs may be highly relevant to human disease. The IMF-mediated mechanism we propose also involves the IBD-associated genes IL1R1, MAPK1, and the PGE2 receptor-encoding PTGER4. Our results establish a previously unidentified myofibroblast-specific innate pathway that regulates intestinal homeostasis and may underlie IBD susceptibility in humans.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Epitelio/metabolismo , Homeostasis , Intestinos/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Colitis/enzimología , Colitis/inmunología , Colitis/patología , Sulfato de Dextran , Dinoprostona/administración & dosificación , Dinoprostona/farmacología , Susceptibilidad a Enfermedades , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/patología , Homeostasis/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/patología , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/patología , Quinasas Quinasa Quinasa PAM/deficiencia , Ratones Endogámicos C57BL , Modelos Biológicos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/enzimología , Miofibroblastos/patología , Fenotipo , Proteínas Proto-Oncogénicas/deficiencia , Transducción de Señal/efectos de los fármacos
10.
J Neurosci ; 34(35): 11684-90, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25164664

RESUMEN

Fever is a hallmark of inflammatory and infectious diseases. The febrile response is triggered by prostaglandin E2 synthesis mediated by induced expression of the enzymes cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase 1 (mPGES-1). The cellular source for pyrogenic PGE2 remains a subject of debate; several hypotheses have been forwarded, including immune cells in the periphery and in the brain, as well as the brain endothelium. Here we generated mice with selective deletion of COX-2 and mPGES1 in brain endothelial cells. These mice displayed strongly attenuated febrile responses to peripheral immune challenge. In contrast, inflammation-induced hypoactivity was unaffected, demonstrating the physiological selectivity of the response to the targeted gene deletions. These findings demonstrate that PGE2 synthesis in brain endothelial cells is critical for inflammation-induced fever.


Asunto(s)
Dinoprostona/biosíntesis , Células Endoteliales/metabolismo , Fiebre/metabolismo , Inflamación/metabolismo , Animales , Ciclooxigenasa 2/metabolismo , Ensayo de Inmunoadsorción Enzimática , Fiebre/etiología , Inmunohistoquímica , Inflamación/complicaciones , Oxidorreductasas Intramoleculares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Prostaglandina-E Sintasas
11.
J Biol Chem ; 289(45): 31647-61, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25262870

RESUMEN

Tissue factor pathway inhibitor-2 (TFPI-2) is a homologue of TFPI-1 and contains three Kunitz-type domains and a basic C terminus region. The N-terminal domain of TFPI-2 is the only inhibitory domain, and it inhibits plasma kallikrein, factor XIa, and plasmin. However, plasma TFPI-2 levels are negligible (≤20 pM) in the context of influencing clotting or fibrinolysis. Here, we report that platelets contain significant amounts of TFPI-2 derived from megakaryocytes. We employed RT-PCR, Western blotting, immunohistochemistry, and confocal microscopy to determine that platelets, MEG-01 megakaryoblastic cells, and bone marrow megakaryocytes contain TFPI-2. ELISA data reveal that TFPI-2 binds factor V (FV) and partially B-domain-deleted FV (FV-1033) with K(d) ~9 nM and binds FVa with K(d) ~100 nM. Steady state analysis of surface plasmon resonance data reveal that TFPI-2 and TFPI-1 bind FV-1033 with K(d) ~36-48 nM and bind FVa with K(d) ~252-456 nM. Further, TFPI-1 (but not TFPI-1161) competes with TFPI-2 in binding to FV. These data indicate that the C-terminal basic region of TFPI-2 is similar to that of TFPI-1 and plays a role in binding to the FV B-domain acidic region. Using pull-down assays and Western blots, we show that TFPI-2 is associated with platelet FV/FVa. TFPI-2 (~7 nM) in plasma of women at the onset of labor is also, in part, associated with FV. Importantly, TFPI-2 in platelets and in plasma of pregnant women inhibits FXIa and tissue-type plasminogen activator-induced clot fibrinolysis. In conclusion, TFPI-2 in platelets from normal or pregnant subjects and in plasma from pregnant women binds FV/Va and regulates intrinsic coagulation and fibrinolysis.


Asunto(s)
Plaquetas/citología , Fibrinólisis/fisiología , Glicoproteínas/metabolismo , Lipoproteínas/metabolismo , Megacariocitos/citología , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Coagulación Sanguínea , Plaquetas/enzimología , Células de la Médula Ósea/citología , Femenino , Sangre Fetal/enzimología , Regulación de la Expresión Génica , Glicoproteínas/genética , Hemostasis , Humanos , Ligandos , Lipoproteínas/genética , Embarazo , Inhibidores de Proteasas/química , Unión Proteica , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
12.
Biochem Biophys Res Commun ; 438(2): 249-56, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23850620

RESUMEN

Cyclooxygenase 2 (COX)-2 is induced by bacterial and viral infections and has complex, poorly understood roles in anti-pathogen immunity. Here, we use a knock-in luciferase reporter model to image Cox2 expression across a range of tissues in mice following treatment with the either the prototypical bacterial pathogen-associated molecular pattern (PAMP), LPS, which activates Toll-like receptor (TLR)4, or with poly(I:C), a viral PAMP, which activates TLR3. LPS induced Cox2 expression in all tissues examined. In contrast, poly(I:C) elicited a milder response, limited to a subset of tissues. A panel of cytokines and interferons was measured in plasma of wild-type, Cox1(-/-) and Cox2(-/-) mice treated with LPS, poly(I:C), MALP2 (TLR2/6), Pam3CSK4 (TLR2/1), R-848 (TLR7/8) or CpG ODN (TLR9), to establish whether/how each COX isoform modulates specific PAMP/TLR responses. Only LPS induced notable loss of condition in mice (inactivity, hunching, piloerection). However, all TLR agonists produced cytokine responses, many of which were modulated in specific fashions by Cox1 or Cox2 gene deletion. Notably we observed opposing effects of Cox2 gene deletion on the responses to the bacterial PAMP, LPS, and the viral PAMP, poly(I:C), consistent with the differing abilities of the PAMPs to induce Cox2 expression. Cox2 gene deletion limited the plasma IL-1ß and interferon-γ responses and hypothermia produced by LPS. In contrast, in response to poly(I:C), Cox2(-/-) mice exhibited enhanced plasma interferon (IFNα,ß,γ,λ) and related cytokine responses (IP-10, IL-12). These observations suggest that a COX-2 selective inhibitor, given early in infection, may enhance and/or prolong endogenous interferon responses, and thereby increase anti-viral immunity.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Interferones/metabolismo , Receptores Toll-Like/metabolismo , Animales , Bacterias/metabolismo , Quimiocina CXCL10/metabolismo , Ciclooxigenasa 2/genética , Femenino , Eliminación de Gen , Técnicas de Sustitución del Gen , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Luciferasas , Masculino , Ratones , Ratones Endogámicos C57BL , Poli I-C/metabolismo , Virus/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(12): 5551-6, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20080663

RESUMEN

Deoxycytidine kinase (dCK) is a rate-limiting enzyme in deoxyribonucleoside salvage, a metabolic pathway that recycles products of DNA degradation. dCK phosphorylates and therefore activates nucleoside analog prodrugs frequently used in cancer, autoimmunity, and viral infections. In contrast to its well established therapeutic relevance, the biological function of dCK remains enigmatic. Highest levels of dCK expression are found in thymus and bone marrow, indicating a possible role in lymphopoiesis. To test this hypothesis we generated and analyzed dCK knockout (KO) mice. dCK inactivation selectively and profoundly affected T and B cell development. A 90-fold decrease in thymic cellularity was observed in the dCK KO mice relative to wild-type littermates. Lymphocyte numbers in the dCK KO mice were 5- to 13-fold below normal values. The severe impact of dCK inactivation on lymphopoiesis was unexpected given that nucleoside salvage has been thought to play a limited, "fine-tuning" role in regulating deoxyribonucleotide triphosphate pools produced by the de novo pathway. The dCK KO phenotype challenges this view and indicates that, in contrast to the great majority of other somatic cells, normal lymphocyte development critically requires the deoxyribonucleoside salvage pathway.


Asunto(s)
Linfocitos B/enzimología , Desoxicitidina Quinasa/fisiología , Linfopoyesis/fisiología , Linfocitos T/enzimología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Desoxicitidina Quinasa/deficiencia , Desoxicitidina Quinasa/genética , Exones , Marcación de Gen , Tejido Linfoide/anomalías , Linfopoyesis/inmunología , Ratones , Ratones Noqueados , Modelos Biológicos , Linfocitos T/citología , Linfocitos T/inmunología
14.
J Biol Chem ; 286(28): 24535-43, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21596747

RESUMEN

Serum coagulation factor X (FX) is proposed to play a major role in adenovirus tropism, promoting transduction by bridging the virus to cell-surface heparan sulfate proteoglycans (HSPGs). Both murine FX and human FX increased transduction by Ad.CMVfLuc, an adenovirus vector, in murine hepatocyte-like cells and human hepatocarcinoma cells. In contrast, only hFX increased transduction of several non-hepatic cancer cell lines and Chinese hamster ovary (CHO) cells. Not only was mFX unable to promote transduction in these cells, it competitively blocked hFX-enhanced transduction. Competition and HSPG digestion experiments suggested mFX- and hFX-enhanced transduction in hepatocyte-derived cells, and hFX-enhanced transduction in epithelial cancer cells were dependent on HSPGs. Ad·hFX-mediated transduction of CHO mutants unable to produce HSPGs was also curtailed. Hepatocyte-derived cells expressed substantially more HSPGs than the cancer cell lines. Dose-response curves and heparin-Sepharose binding suggested Ad·hFX has greater affinity for HSPGs than does Ad·mFX. In coagulation factor-depleted mice hFX also had enhanced ability, compared with mFX, to reconstitute hepatic adenovirus transduction. The results suggest that differences in Ad·hFX and Ad·mFX affinity to HSPGs may result in differences in their ability to enhance adenovirus transduction of many cells. These findings may have implications for murine models of adenovirus vector targeting.


Asunto(s)
Adenoviridae/fisiología , Factor X/farmacología , Heparitina Sulfato/metabolismo , Proteoglicanos/metabolismo , Transducción Genética/métodos , Tropismo Viral/efectos de los fármacos , Animales , Células CHO , Cricetinae , Cricetulus , Factor X/química , Vectores Genéticos/metabolismo , Células Hep G2 , Heparitina Sulfato/química , Humanos , Ratones , Mutación , Especificidad de Órganos , Proteoglicanos/química , Especificidad de la Especie , Tropismo Viral/fisiología
15.
Proc Natl Acad Sci U S A ; 106(8): 2847-52, 2009 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-19196993

RESUMEN

Gemcitabine (2',2'-difluorodeoxycytidine, dFdC) and cytosine arabinoside (cytarabine, ara-C) represent a class of nucleoside analogs used in cancer chemotherapy. Administered as prodrugs, dFdC and ara-C are transported across cell membranes and are converted to cytotoxic derivatives through consecutive phosphorylation steps catalyzed by endogenous nucleoside kinases. Deoxycytidine kinase (DCK) controls the rate-limiting step in the activation cascade of dFdC and ara-C. DCK activity varies significantly among individuals and across different tumor types and is a critical determinant of tumor responses to these prodrugs. Current assays to measure DCK expression and activity require biopsy samples and are prone to sampling errors. Noninvasive methods that can detect DCK activity in tumor lesions throughout the body could circumvent these limitations. Here, we demonstrate an approach to detecting DCK activity in vivo by using positron emission tomography (PET) and (18)F-labeled 1-(2'-deoxy-2'-fluoroarabinofuranosyl) cytosine] ([(18)F]FAC), a PET probe recently developed by our group. We show that [(18)F]FAC is a DCK substrate with an affinity similar to that of dFdC. In vitro, accumulation of [(18)F]FAC in murine and human leukemia cell lines is critically dependent on DCK activity and correlates with dFdC sensitivity. In mice, [(18)F]FAC accumulates selectively in DCK-positive vs. DCK-negative tumors, and [(18)F]FAC microPET scans can predict responses to dFdC. We suggest that [(18)F]FAC PET might be useful for guiding treatment decisions in certain cancers by enabling individualized chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Leucemia/diagnóstico por imagen , Animales , Antineoplásicos/farmacocinética , Citosina/análogos & derivados , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Humanos , Leucemia/tratamiento farmacológico , Ratones , Tomografía de Emisión de Positrones , Gemcitabina
16.
Proc Natl Acad Sci U S A ; 106(51): 21984-9, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19995983

RESUMEN

Identifying the molecular targets for the beneficial or detrimental effects of small-molecule drugs is an important and currently unmet challenge. We have developed a method, drug affinity responsive target stability (DARTS), which takes advantage of a reduction in the protease susceptibility of the target protein upon drug binding. DARTS is universally applicable because it requires no modification of the drug and is independent of the mechanism of drug action. We demonstrate use of DARTS to identify known small-molecule-protein interactions and to reveal the eukaryotic translation initiation machinery as a molecular target for the longevity-enhancing plant natural product resveratrol. We envisage that DARTS will also be useful in global mapping of protein-metabolite interaction networks and in label-free screening of unlimited varieties of compounds for development as molecular imaging agents.


Asunto(s)
Sistemas de Liberación de Medicamentos , Sitios de Unión , ADN Complementario , Proteómica , Resveratrol , Estilbenos/metabolismo
17.
Cell Mol Gastroenterol Hepatol ; 13(4): 1095-1120, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35017061

RESUMEN

BACKGROUND AND AIMS: Phagocytosis (efferocytosis) of apoptotic neutrophils by macrophages anchors the resolution of intestinal inflammation. Efferocytosis prevents secondary necrosis and inhibits further inflammation, and also reprograms macrophages to facilitate tissue repair and promote resolution function. Macrophage efferocytosis and efferocytosis-dependent reprogramming are implicated in the pathogenesis of inflammatory bowel disease. We previously reported that absence of macrophage cyclooxygenase 2 (COX2) exacerbates inflammatory bowel disease-like intestinal inflammation. To elucidate the underlying pathogenic mechanism, we investigated here whether COX2 mediates macrophage efferocytosis and efferocytosis-dependent reprogramming, including intestinal epithelial repair capacity. METHODS: Using apoptotic neutrophils and synthetic apoptotic targets, we determined the effects of macrophage specific Cox2 knockout and pharmacological COX2 inhibition on the efferocytosis capacity of mouse primary macrophages. COX2-mediated efferocytosis-dependent eicosanoid lipidomics was determined by liquid chromatography tandem mass spectrometry. Small intestinal epithelial organoids were employed to assay the effects of COX2 on efferocytosis-dependent intestinal epithelial repair. RESULTS: Loss of COX2 impaired efferocytosis in mouse primary macrophages, in part, by affecting the binding capacity of macrophages for apoptotic cells. This effect was comparable to that of high-dose lipopolysaccharide and was accompanied by both dysregulation of macrophage polarization and the inhibited expression of genes involved in apoptotic cell binding. COX2 modulated the production of efferocytosis-dependent lipid inflammatory mediators that include the eicosanoids prostaglandin I2, prostaglandin E2, lipoxin A4, and 15d-PGJ2; and further affected secondary efferocytosis. Finally, macrophage efferocytosis induced, in a macrophage COX2-dependent manner, a tissue restitution and repair phenotype in intestinal epithelial organoids. CONCLUSIONS: Macrophage COX2 potentiates efferocytosis capacity and efferocytosis-dependent reprogramming, facilitating macrophage intestinal epithelial repair capacity.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Enfermedades Inflamatorias del Intestino , Fagocitosis , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/farmacología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , Macrófagos/metabolismo , Ratones , Fagocitosis/genética
18.
Carcinogenesis ; 32(3): 417-26, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21156970

RESUMEN

Patients with inflammatory bowel diseases are at increased risk for colorectal cancer. Pharmacological inhibition of cyclooxygenase (COX) function exacerbates symptoms in colitis patients. Animal models of colitis using Cox-2-knockout mice and COX inhibitors also indicate that COX-2 has a protective role against colon inflammation. However, because conventional Cox-2 deletion and COX-2 inhibitors eliminate COX-2 function in all cells, it has not been possible to analyze the role(s) of COX-2 in different cell types. Here, we use a Cox-2(flox) conditional knockout mouse to analyze the role of COX-2 expression in distinct cell types in the colon in response to dextran sulfate sodium (DSS)-induced colitis. We generated Cox-2 conditional knockouts in myeloid cells with LysMCre knock-in mice, in endothelial cells with VECadCreERT2 transgenic mice and in epithelial cells with VillinCre transgenic mice. When treated with DSS to induce colitis, both myeloid cell-specific and endothelial cell-specific Cox-2-knockout mice exhibited greater weight loss, increased clinical scores and decreased epithelial cell proliferation after DSS injury when compared with littermate controls. In contrast, epithelial-specific Cox-2 knockouts and control littermates did not differ in response to DSS. These results suggest that COX-2 expression in myeloid cells and endothelial cells, but not epithelial cells, is important for protection of epithelial cells in this murine colitis model.


Asunto(s)
Colitis/enzimología , Ciclooxigenasa 2/fisiología , Células Endoteliales/enzimología , Células Epiteliales/enzimología , Células Mieloides/enzimología , Animales , Apoptosis , Southern Blotting , Western Blotting , Proliferación Celular , Colitis/etiología , Colitis/patología , Colon/citología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Hígado/citología , Luciferasas/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/efectos de los fármacos
19.
Arterioscler Thromb Vasc Biol ; 30(2): 260-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19926832

RESUMEN

OBJECTIVE: The role of myeloid cell cyclooxygenase-2 (COX-2) in the progression of atherosclerosis has not been clearly defined. METHODS AND RESULTS: We investigated the role of COX-2 expressed in the myeloid lineage in the development of atherosclerosis using a myeloid-specific COX-2(-/-) (COX-2(-M/-M)) mouse on a hyperlipidemic apolipoprotein (apo) E(-/-) background (COX-2(-M/-M)/apoE(-/-)). Myeloid COX-2 depletion resulted in significant attenuation of acute inflammation corresponding with decreased PGE(2) levels in an air pouch model. COX-2 depletion in myeloid cells did not influence development of atherosclerosis in COX-2(-M/-M)/apoE(-/-) when compared to apoE(-/-) littermates fed either chow or western diets. The unanticipated lack of contribution of myeloid COX-2 to the development atherosclerosis is not attributable to altered maintenance, differentiation, or mobilization of myeloid and lymphoid populations. Moreover, myeloid COX-2 depletion resulted in unaltered serum prostanoid levels and cellular composition of atherosclerotic lesions of COX-2(-M/-M)/apoE(-/-) mice. CONCLUSIONS: Our results suggest that COX-2 expression in myeloid cells, including macrophages, does not influence the development of atherosclerosis in mice.


Asunto(s)
Enfermedades de la Aorta/enzimología , Apolipoproteínas E/deficiencia , Aterosclerosis/enzimología , Ciclooxigenasa 2/deficiencia , Inflamación/enzimología , Células Mieloides/enzimología , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Células Cultivadas , Ciclooxigenasa 2/genética , Grasas de la Dieta , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/patología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Sci Adv ; 7(12)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33741600

RESUMEN

Endothelial cyclooxygenase-1-derived prostanoids, including prostacyclin, have clear cardioprotective roles associated with their anti-thrombotic potential but have also been suggested to have paradoxical pathological activities within arteries. To date it has not been possible to test the importance of this because no models have been available that separate vascular cyclooxygenase-1 products from those generated elsewhere. Here, we have used unique endothelial-specific cyclooxygenase-1 knockout mice to show that endothelial cyclooxygenase-1 produces both protective and pathological products. Functionally, however, the overall effect of these was to drive pathological responses in the context of both vasoconstriction in vitro and the development of atherosclerosis and vascular inflammation in vivo. These data provide the first demonstration of a pathological role for the vascular cyclooxygenase-1 pathway, highlighting its potential as a therapeutic target. They also emphasize that, across biology, the role of prostanoids is not always predictable due to unique balances of context, products, and receptors.


Asunto(s)
Aterosclerosis , Ciclooxigenasa 1/metabolismo , Epoprostenol , Proteínas de la Membrana/metabolismo , Animales , Aterosclerosis/etiología , Ciclooxigenasa 1/genética , Epoprostenol/metabolismo , Ratones , Prostaglandinas , Vasoconstricción
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda