Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
ACS Med Chem Lett ; 12(11): 1832-1839, 2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34795874

RESUMEN

Calcium and integrin binding protein 1 (CIB1) is a small, intracellular protein recently implicated in survival and proliferation of triple-negative breast cancer (TNBC). Considering its interactions with PAK1 and downstream signaling, CIB1 has been suggested as a potential therapeutic target in TNBC. As such, CIB1 has been the focus of inhibitor discovery efforts. To overcome issues of potency and stability in previously reported CIB1 inhibitors, we deploy mRNA display to discover new cyclic peptide inhibitors with improved biophysical properties and cellular activity. We advance UNC10245131, a cyclic peptide with low nanomolar affinity and good selectivity for CIB1 over other EF-hand domain proteins and improved permeability and stability over previously identified linear peptide inhibitor UNC10245092. Unlike UNC10245092, UNC10245131 lacks cytotoxicity and does not affect downstream signaling. Despite this, UNC10245131 is a potent ligand that could aid in clarifying roles of CIB1 in TNBC survival and proliferation and other CIB1-associated biological phenotypes.

2.
ACS Chem Biol ; 15(6): 1505-1516, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32383857

RESUMEN

Calcium and integrin binding protein 1 (CIB1) is an EF-hand-containing, small intracellular protein that has recently been implicated in cancer cell survival and proliferation. In particular, CIB1 depletion significantly impairs tumor growth in triple-negative breast cancer (TNBC). Thus, CIB1 is a potentially attractive target for cancer chemotherapy that has yet to be validated by a chemical probe. To produce a probe molecule to the CIB1 helix 10 (H10) pocket and demonstrate that it is a viable target for molecular intervention, we employed random peptide phage display to screen and select CIB1-binding peptides. The top peptide sequence selected, UNC10245092, was produced synthetically, and binding to CIB1 was confirmed by isothermal titration calorimetry (ITC) and a time-resolved fluorescence resonance energy transfer (TR-FRET) assay. Both assays showed that the peptide bound to CIB1 with low nanomolar affinity. CIB1 was cocrystallized with UNC10245092, and the 2.1 Å resolution structure revealed that the peptide binds as an α-helix in the H10 pocket, displacing the CIB1 C-terminal H10 helix and causing conformational changes in H7 and H8. UNC10245092 was further derivatized with a C-terminal Tat-derived cell penetrating peptide (CPP) to demonstrate its effects on TNBC cells in culture, which are consistent with results of CIB1 depletion. These studies provide a first-in-class chemical tool for CIB1 inhibition in cell culture and validate the CIB1 H10 pocket for future probe and drug discovery efforts.


Asunto(s)
Proteínas de Unión al Calcio/antagonistas & inhibidores , Secuencia de Aminoácidos , Calorimetría/métodos , Línea Celular Tumoral , Descubrimiento de Drogas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Conformación Proteica
3.
Blood Adv ; 3(22): 3818-3828, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31770438

RESUMEN

We previously reported the discovery of a novel lipid deacetylase in platelets, arylacetamide deacetylase-like 1 (AADACL1/NCEH1), and that its inhibition impairs agonist-induced platelet aggregation, Rap1 GTP loading, protein kinase C (PKC) activation, and ex vivo thrombus growth. However, precise mechanisms by which AADACL1 impacts platelet signaling and function in vivo are currently unknown. Here, we demonstrate that AADACL1 regulates the accumulation of ether lipids that impact PKC signaling networks crucial for platelet activation in vitro and in vivo. Human platelets treated with the AADACL1 inhibitor JW480 or the AADACL1 substrate 1-O-hexadecyl-2-acetyl-sn-glycerol (HAG) exhibited decreased platelet aggregation, granule secretion, Ca2+ flux, and PKC phosphorylation. Decreased aggregation and secretion were rescued by exogenous adenosine 5'-diphosphate, indicating that AADACL1 likely functions to induce dense granule secretion. Experiments with P2Y12-/- and CalDAG GEFI-/- mice revealed that the P2Y12 pathway is the predominate target of HAG-mediated inhibition of platelet aggregation. HAG itself displayed weak agonist properties and likely mediates its inhibitory effects via conversion to a phosphorylated metabolite, HAGP, which directly interacted with the C1a domains of 2 distinct PKC isoforms and blocked PKC kinase activity in vitro. Finally, AADACL1 inhibition in rats reduced platelet aggregation, protected against FeCl3-induced arterial thrombosis, and delayed tail bleeding time. In summary, our data support a model whereby AADACL1 inhibition shifts the platelet ether lipidome to an inhibitory axis of HAGP accumulation that impairs PKC activation, granule secretion, and recruitment of platelets to sites of vascular damage.


Asunto(s)
Plaquetas/metabolismo , Metabolismo de los Lípidos , Esterol Esterasa/metabolismo , Trombosis/etiología , Trombosis/metabolismo , Animales , Plaquetas/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Modelos Biológicos , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Unión Proteica , Proteína Quinasa C/metabolismo , Ratas , Receptores Purinérgicos P2Y12/metabolismo , Transducción de Señal/efectos de los fármacos , Esterol Esterasa/antagonistas & inhibidores , Especificidad por Sustrato , Trombosis/tratamiento farmacológico
4.
Adv Sci (Weinh) ; 6(21): 1900582, 2019 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-31728272

RESUMEN

Delivery of nucleic acids into solid tumor environments remains a pressing challenge. This study examines the ability of macrophages to horizontally transfer small interfering RNA (siRNA) lipoplexes to cancer cells. Macrophages are a natural candidate for a drug carrier because of their ability to accumulate at high densities into many cancer types, including, breast, prostate, brain, and colon cancer. Here, it is demonstrated that macrophages can horizontally transfer siRNA to cancer cells during in vitro coculture. The amount of transfer can be dosed depending on the amount of siRNA loaded and total number of macrophages delivered. Macrophages loaded with calcium integrin binding protein-1 (CIB1)-siRNA result in decreased tumorsphere growth and decreased mRNA expression of CIB1 and KI67 in MDA-MB-468 human breast cancer cells. Adoptive transfer of macrophages transfected with CIB1-siRNA localizes to the orthotopic MDA-MB-468 tumor. Furthermore, it is reported that macrophage activation can modulate this transfer process as well as intracellular trafficking protein Rab27a. As macrophages are heavily involved in tumor progression, understanding how to use macrophages for drug delivery can substantially benefit the treatment of tumors.

5.
Cancer Cell Int ; 19: 26, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30740034

RESUMEN

BACKGROUND: Patients diagnosed with triple negative breast cancer (TNBC) have limited treatment options and often suffer from resistance and toxicity due to chemotherapy. We previously found that depleting calcium and integrin-binding protein 1 (CIB1) induces cell death selectively in TNBC cells, while sparing normal cells. Therefore, we asked whether CIB1 depletion further enhances tumor-specific killing when combined with either the commonly used chemotherapeutic, docetaxel, or the cell death-inducing ligand, TRAIL. METHODS: We targeted CIB1 by RNA interference in MDA-MB-436, MDA-MB-231, MDA-MB-468, docetaxel-resistant MDA-MB-436 TNBC cells and ME16C normal breast epithelial cells alone or combination with docetaxel or TRAIL. Cell death was quantified via trypan blue exclusion using flow cytometry and cell death mechanisms were analyzed by Western blotting. Cell surface levels of TRAIL receptors were measured by flow cytometry analysis. RESULTS: CIB1 depletion combined with docetaxel significantly enhanced tumor-specific cell death relative to each treatment alone. The enhanced cell death strongly correlated with caspase-8 activation, a hallmark of death receptor-mediated apoptosis. The death receptor TRAIL-R2 was upregulated in response to CIB1 depletion, which sensitized TNBC cells to the ligand TRAIL, resulting in a synergistic increase in cell death. In addition to death receptor-mediated apoptosis, both combination treatments activated a non-apoptotic mechanism, called paraptosis. Interestingly, these combination treatments also induced nearly complete death of docetaxel-resistant MDA-MB-436 cells, again via apoptosis and paraptosis. In contrast, neither combination treatment induced cell death in normal ME16C cells. CONCLUSION: Novel combinations of CIB1 depletion with docetaxel or TRAIL selectively enhance naive and docetaxel-resistant TNBC cell death while sparing normal cell. Therefore, combination therapies that target CIB1 could prove to be a safe and durable strategy for treatment of TNBC and potentially other cancers.

6.
J Exp Med ; 215(9): 2289-2310, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30068544

RESUMEN

Patients with epidermodysplasia verruciformis (EV) and biallelic null mutations of TMC6 (encoding EVER1) or TMC8 (EVER2) are selectively prone to disseminated skin lesions due to keratinocyte-tropic human ß-papillomaviruses (ß-HPVs), which lack E5 and E8. We describe EV patients homozygous for null mutations of the CIB1 gene encoding calcium- and integrin-binding protein-1 (CIB1). CIB1 is strongly expressed in the skin and cultured keratinocytes of controls but not in those of patients. CIB1 forms a complex with EVER1 and EVER2, and CIB1 proteins are not expressed in EVER1- or EVER2-deficient cells. The known functions of EVER1 and EVER2 in human keratinocytes are not dependent on CIB1, and CIB1 deficiency does not impair keratinocyte adhesion or migration. In keratinocytes, the CIB1 protein interacts with the HPV E5 and E8 proteins encoded by α-HPV16 and γ-HPV4, respectively, suggesting that this protein acts as a restriction factor against HPVs. Collectively, these findings suggest that the disruption of CIB1-EVER1-EVER2-dependent keratinocyte-intrinsic immunity underlies the selective susceptibility to ß-HPVs of EV patients.


Asunto(s)
Betapapillomavirus/inmunología , Proteínas de Unión al Calcio/inmunología , Epidermodisplasia Verruciforme/inmunología , Inmunidad Innata , Queratinocitos/inmunología , Proteínas de la Membrana/inmunología , Complejos Multiproteicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Adhesión Celular/inmunología , Movimiento Celular/inmunología , Epidermodisplasia Verruciforme/patología , Femenino , Papillomavirus Humano 16/inmunología , Humanos , Queratinocitos/patología , Masculino , Persona de Mediana Edad , Proteínas Oncogénicas Virales/inmunología
8.
Sci Rep ; 7(1): 12178, 2017 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-28939911

RESUMEN

Calcium and integrin binding protein 1 (CIB1) is a calcium-binding protein that was initially identified as a binding partner of platelet integrin αIIb. Although CIB1 has been shown to interact with multiple proteins, its biological function in the brain remains unclear. Here, we show that CIB1 negatively regulates degeneration of dopaminergic neurons in a mouse model of Parkinson's disease using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Genetic deficiency of the CIB1 gene enhances MPTP-induced neurotoxicity in dopaminergic neurons in CIB1-/- mice. Furthermore, RNAi-mediated depletion of CIB1 in primary dopaminergic neurons potentiated 1-methyl-4-phenyl pyrinidium (MPP+)-induced neuronal death. CIB1 physically associated with apoptosis signal-regulating kinase 1 (ASK1) and thereby inhibited the MPP+-induced stimulation of the ASK1-mediated signaling cascade. These findings suggest that CIB1 plays a protective role in MPTP/MPP+-induced neurotoxicity by blocking ASK1-mediated signaling.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Intoxicación por MPTP/patología , Enfermedad de Parkinson/patología , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Apoptosis/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
9.
Physiol Rep ; 5(5)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28275112

RESUMEN

We generated and studied CLIC1 null (C1KO) mice to investigate the physiological role of this protein. C1KO and matched wild-type (WT) mice were studied in two models of acute toxic tissue injury. CLIC1 expression is upregulated following acute injury of WT kidney and pancreas and is absent in C1KOs. Acute tissue injury is attenuated in the C1KOs and this correlates with an absence of the rise in tissue reactive oxygen species (ROS) that is seen in WT mice. Infiltration of injured tissue by inflammatory cells was comparable between WT and C1KOs. Absence of CLIC1 increased PMA-induced superoxide production by isolated peritoneal neutrophils but dramatically decreased PMA-induced superoxide production by peritoneal macrophages. CLIC1 is expressed in both neutrophils and macrophages in a peripheral pattern consistent with either plasma membrane or the cortical cytoskeleton in resting cells and redistributes away from the periphery following PMA stimulation in both cell types. Absence of CLIC1 had no effect on redistribution or dephosphorylation of Ezrin/ERM cytoskeleton in macrophages. Plasma membrane chloride conductance is altered in the absence of CLIC1, but not in a way that would be expected to block superoxide production. NADPH oxidase redistributes from an intracellular compartment to the plasma membrane when WT macrophages are stimulated to produce superoxide and this redistribution fails to occur in C1KO macrophages. We conclude that the role of CLIC1 in macrophage superoxide production is to support redistribution of NADPH oxidase to the plasma membrane, and not through major effects on ERM cytoskeleton or by acting as a plasma membrane chloride channel.


Asunto(s)
Lesión Renal Aguda/metabolismo , Canales de Cloruro/metabolismo , Macrófagos/metabolismo , Superóxidos/metabolismo , Lesión Renal Aguda/genética , Animales , Membrana Celular/metabolismo , Canales de Cloruro/genética , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasas/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo
10.
FASEB J ; 30(8): 2640-50, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27118676

RESUMEN

Calcium- and integrin-binding protein 1 (CIB1) is a small, ubiquitously expressed protein that was first identified as an intracellular binding partner of a platelet-specific α-integrin cytoplasmic tail. Although early studies revealed a role for CIB1 in regulating platelet integrin activity, recent studies have indicated a more diverse role for CIB1 in many different cell types and processes, including calcium signaling, migration, adhesion, proliferation, and survival. Increasing evidence also points to a novel role for CIB1 in cancer and cardiovascular disease. In addition, an array of CIB1 binding partners has been identified that provide important insight into how CIB1 may regulate these processes. Some of these binding partners include the serine/threonine kinases, p21-activated kinase 1 (PAK1), apoptosis signal-regulating kinase 1 (ASK1), and polo-like kinase 3 (PLK3). Structural and mutational studies indicate that CIB1 binds most or all of its partners via a well-defined hydrophobic cleft. Although CIB1 itself lacks known enzymatic activity, it supports the PI3K/AKT and MEK/ERK oncogenic signaling pathways, in part, by directly modulating enzymes in these pathways. In this review, we discuss our current understanding of CIB1 and key questions regarding structure and function and how this seemingly diminutive protein impacts important signaling pathways and cellular processes in human health and disease.-Leisner, T. M., Freeman, T. C., Black, J. L., Parise, L. V. CIB1: a small protein with big ambitions.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de Unión al Calcio/genética , Enfermedades Cardiovasculares/metabolismo , Humanos , Neoplasias/metabolismo
11.
Angew Chem Int Ed Engl ; 55(3): 950-4, 2016 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-26636264

RESUMEN

Although peptide-based therapeutics are finding increasing application in the clinic, extensive structural modification is typically required to prevent their rapid degradation by proteases in the blood. We have evaluated the ability of erythrocytes to serve as reservoirs, protective shields (against proteases), and light-triggered launch pads for peptides. We designed lipidated peptides that are anchored to the surface of red blood cells, which furnishes a protease-resistant environment. A photocleavable moiety is inserted between the lipid anchor and the peptide backbone, thereby enabling light-triggered peptide release from erythrocytes. We have shown that a cell-permeable peptide, a hormone (melanocyte stimulating hormone), and a blood-clotting agent can be anchored to erythrocytes, protected from proteases, and photolytically released to create the desired biological effect.


Asunto(s)
Membrana Celular/efectos de los fármacos , Proteínas de la Membrana/química , Péptidos/uso terapéutico , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Péptidos/química
12.
Breast Cancer Res Treat ; 152(2): 337-46, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26105795

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with generally poor prognosis and no available targeted therapies, highlighting a critical unmet need to identify and characterize novel therapeutic targets. We previously demonstrated that CIB1 is necessary for cancer cell survival and proliferation via regulation of two oncogenic signaling pathways, RAF-MEK-ERK and PI3K-AKT. Because these pathways are often upregulated in TNBC, we hypothesized that CIB1 may play a broader role in TNBC cell survival and tumor growth. Methods utilized include inducible RNAi depletion of CIB1 in vitro and in vivo, immunoblotting, clonogenic assay, flow cytometry, RNA-sequencing, bioinformatics analysis, and Kaplan-Meier survival analysis. CIB1 depletion resulted in significant cell death in 8 of 11 TNBC cell lines tested. Analysis of components related to PI3K-AKT and RAF-MEK-ERK signaling revealed that elevated AKT activation status and low PTEN expression were key predictors of sensitivity to CIB1 depletion. Furthermore, CIB1 knockdown caused dramatic shrinkage of MDA-MB-468 xenograft tumors in vivo. RNA sequence analysis also showed that CIB1 depletion in TNBC cells activates gene programs associated with decreased proliferation and increased cell death. CIB1 expression levels per se did not predict TNBC susceptibility to CIB1 depletion, and CIB1 mRNA expression levels did not associate with TNBC patient survival. Our data are consistent with the emerging concept of non-oncogene addiction, where a large subset of TNBCs depend on CIB1 for cell survival and tumor growth, independent of CIB1 expression levels. Our data establish CIB1 as a novel therapeutic target for TNBC.


Asunto(s)
Proteínas de Unión al Calcio/genética , Supervivencia Celular/genética , Neoplasias de la Mama Triple Negativas/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Pronóstico , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral , Quinasas p21 Activadas/metabolismo
13.
Chem Biol ; 20(9): 1125-34, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23993462

RESUMEN

A comprehensive knowledge of the platelet proteome is necessary for understanding thrombosis and for envisioning antiplatelet therapies. To discover other biochemical pathways in human platelets, we screened platelets with a carbamate library designed to interrogate the serine hydrolase subproteome and used competitive activity-based protein profiling to map the targets of active carbamates. We identified an inhibitor that targets arylacetamide deacetylase-like 1 (AADACL1), a lipid deacetylase originally identified in invasive cancers. Using this compound, along with highly selective second-generation inhibitors of AADACL1, metabolomics, and RNA interference, we show that AADACL1 regulates platelet aggregation, thrombus growth, RAP1 and PKC activation, lipid metabolism, and fibrinogen binding to platelets and megakaryocytes. These data provide evidence that AADACL1 regulates platelet and megakaryocyte activation and highlight the value of this chemoproteomic strategy for target discovery in platelets.


Asunto(s)
Plaquetas/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Carbamatos/química , Carbamatos/metabolismo , Carbamatos/farmacología , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Línea Celular , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Fibrinógeno/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Metabolómica , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Proteína Quinasa C/metabolismo , Proteómica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Esterol Esterasa , Proteínas de Unión al GTP rap1/metabolismo
14.
Thromb Res ; 132(3): 341-5, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23973010

RESUMEN

INTRODUCTION: The contribution of platelet activation to the pathogenesis of sickle cell disease (SCD) remains uncertain. We evaluated the safety and efficacy of eptifibatide, a synthetic peptide inhibitor of the αIIbß3 receptor, in SCD patients during acute painful episodes. MATERIALS AND METHODS: In this single site, double-blind, placebo-controlled trial, eligible patients with SCD admitted for acute painful episodes were randomized to receive eptifibatide or placebo at a ratio of 2:1. RESULTS: Thirteen patients (SS - 10, Sß(0) - 2, SC - 1) were randomized to receive either eptifibatide (N=9; 6 females; median age - 25years) or placebo (N=4; 3 females; median age - 31years). In the intent-to-treat analysis, there were no major bleeding episodes in either the eptifibatide or placebo arms (point estimate of difference: 0.00, 95% CI; -0.604, 0.372). There was one minor bleeding episode in the eptifibatide arm (point estimate of difference for any bleeding: 0.11, 95% CI: -0.502, 0.494). There was no significant difference in the proportion of patients with thrombocytopenia between the treatment groups (point estimate of difference: 0.11, 95% CI: -0.587, 0.495). There were no differences in the median times to discharge, median times to crisis resolution or the median total opioid use. CONCLUSIONS: In this small study, eptifibatide appeared to be safe, but did not improve the times to crisis resolution or hospital discharge. Adequately powered studies are required to evaluate the safety and efficacy of eptifibatide in SCD. Clinicaltrials.gov Identifier: NCT00834899.


Asunto(s)
Dolor Agudo/tratamiento farmacológico , Anemia de Células Falciformes/complicaciones , Péptidos/uso terapéutico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Dolor Agudo/sangre , Dolor Agudo/etiología , Adolescente , Adulto , Anemia de Células Falciformes/sangre , Método Doble Ciego , Eptifibatida , Femenino , Humanos , Masculino , Persona de Mediana Edad , Péptidos/efectos adversos , Proyectos Piloto , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/efectos adversos , Resultado del Tratamiento , Adulto Joven
15.
Cell Rep ; 3(3): 678-88, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23434512

RESUMEN

MyD88, the intracellular adaptor of most TLRs, mediates either proinflammatory or immunosuppressive signaling that contributes to chronic inflammation-associated diseases. Although gene-specific chromatin modifications regulate inflammation, the role of MyD88 signaling in establishing such epigenetic landscapes under different inflammatory states remains elusive. Using quantitative proteomics to enumerate the inflammation-phenotypic constituents of the MyD88 interactome, we found that in endotoxin-tolerant macrophages, protein phosphatase 2A catalytic subunit α (PP2Ac) enhances its association with MyD88 and is constitutively activated. Knockdown of PP2Ac prevents suppression of proinflammatory genes and resistance to apoptosis. Through site-specific dephosphorylation, constitutively active PP2Ac disrupts the signal-promoting TLR4-MyD88 complex and broadly suppresses the activities of multiple proinflammatory/proapoptotic pathways as well, shifting proinflammatory MyD88 signaling to a prosurvival mode. Constitutively active PP2Ac translocated with MyD88 into the nuclei of tolerant macrophages establishes the immunosuppressive pattern of chromatin modifications and represses chromatin remodeling to selectively silence proinflammatory genes, coordinating the MyD88-dependent inflammation control at both signaling and epigenetic levels under endotoxin-tolerant conditions.


Asunto(s)
Epigénesis Genética/inmunología , Tolerancia Inmunológica/genética , Lipopolisacáridos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína Fosfatasa 2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Apoptosis , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Células HEK293 , Humanos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Fenotipo , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/genética , Proteoma/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
16.
Blood ; 120(3): 636-46, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22661702

RESUMEN

Sickle cell disease (SCD) is associated with a complex vascular pathophysiology that includes activation of coagulation and inflammation. However, the crosstalk between these 2 systems in SCD has not been investigated. Here, we examined the role of tissue factor (TF) in the activation of coagulation and inflammation in 2 different mouse models of SCD (BERK and Townes). Leukocytes isolated from BERK mice expressed TF protein and had increased TF activity compared with control mice. We found that an inhibitory anti-TF antibody abrogated the activation of coagulation but had no effect on hemolysis or anemia. Importantly, inhibition of TF also attenuated inflammation and endothelial cell injury as demonstrated by reduced plasma levels of IL-6, serum amyloid P, and soluble vascular cell adhesion molecule-1. In addition, we found decreased levels of the chemokines MCP-1 and KC, as well as myeloperoxidase in the lungs of sickle cell mice treated with the anti-TF antibody. Finally, we found that endothelial cell-specific deletion of TF had no effect on coagulation but selectively attenuated plasma levels of IL-6. Our data indicate that different cellular sources of TF contribute to activation of coagulation, vascular inflammation, and endothelial cell injury. Furthermore, it appears that TF contributes to these processes without affecting intravascular hemolysis.


Asunto(s)
Anemia de Células Falciformes/inmunología , Coagulación Sanguínea/fisiología , Inflamación/inmunología , Tromboplastina/inmunología , Tromboplastina/metabolismo , Anemia de Células Falciformes/sangre , Animales , Quimiocina CCL2/sangre , Quimiocina CXCL1/sangre , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Eritrocitos/citología , Eritrocitos/fisiología , Femenino , Hemólisis/fisiología , Inflamación/sangre , Interleucina-6/sangre , Leucocitos/inmunología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Neutrófilos/inmunología , Componente Amiloide P Sérico/metabolismo , Molécula 1 de Adhesión Celular Vascular/sangre
17.
Curr Drug Targets ; 12(12): 1859-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21718236

RESUMEN

Platelets are dynamic blood cells that form life-threatening thrombi in response to a variety of pathological conditions such as atherosclerosis, diabetes, metastatic cancer, sickle cell disease and obesity. These thrombi can lead directly to myocardial infarction (MI), stroke and other thrombotic events that contribute to over a million deaths every year in the United States. Even though multiple, effective drugs have been developed to combat these pathologies by antagonizing platelet receptors and their ligands, clinical use of these drugs can result in serious bleeding consequences. With the advent of increasingly powerful and accessible systems biology approaches, however, new opportunities are available to identify novel platelet targets and elucidate potentially safer antiplatelet strategies. Here we provide an overview of some of these exciting systems approaches ranging from genomics to aptamer discovery and emphasize the importance of identifying and exploiting novel platelet targets for therapeutic benefit.


Asunto(s)
Plaquetas/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Biología de Sistemas/métodos , Trombosis/prevención & control , Animales , Plaquetas/efectos de los fármacos , Descubrimiento de Drogas/tendencias , Monitoreo de Drogas , Humanos , Terapia Molecular Dirigida/tendencias , Farmacogenética/tendencias , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Proteómica/tendencias , Medición de Riesgo/tendencias , Transducción de Señal/efectos de los fármacos , Trombosis/tratamiento farmacológico , Trombosis/etiología , Investigación Biomédica Traslacional
18.
J Angiogenes Res ; 2: 17, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20804551

RESUMEN

BACKGROUND: Pathological angiogenesis contributes to various ocular, malignant, and inflammatory disorders, emphasizing the need to understand this process more precisely on a molecular level. Previously we found that CIB1, a 22 kDa regulatory protein, plays a critical role in endothelial cell function, angiogenic growth factor-mediated cellular functions, PAK1 activation, MMP-2 expression, and in vivo ischemia-induced angiogenesis. Since pathological angiogenesis is highly dependent on many of these same processes, we hypothesized that CIB1 may also regulate tumor-induced angiogenesis. METHODS: To test this hypothesis, we allografted either murine B16 melanoma or Lewis lung carcinoma cells into WT and CIB1-KO mice, and monitored tumor growth, morphology, histology, and intra-tumoral microvessel density. RESULTS: Allografted melanoma tumors that developed in CIB1-KO mice were smaller in volume, had a distinct necrotic appearance, and had significantly less intra-tumoral microvessel density. Similarly, allografted Lewis lung carcinoma tumors in CIB1-KO mice were smaller in volume and mass, and appeared to have decreased perfusion. Intra-tumoral hemorrhage, necrosis, and perivascular fibrosis were also increased in tumors that developed in CIB1-KO mice. CONCLUSIONS: These findings suggest that, in addition to its other functions, CIB1 plays a critical role in facilitating tumor growth and tumor-induced angiogenesis.

19.
Curr Top Dev Biol ; 80: 311-35, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17950378

RESUMEN

Proper platelet function is essential for hemostasis. However, understanding platelet function is complicated by the fact that platelets are anucleate and therefore not amenable to direct genetic manipulations. To study platelet function, several laboratories have developed CHO cell lines expressing platelet proteins or used megakaryocyte-like cell lines. However, these cell culture models are unable to mimic critical platelet functions, most notably agonist-induced activation of integrin alphaIIbbeta3. Mature megakaryocytes, which are platelet precursors, express platelet-specific proteins, and the function of such proteins and signaling pathways appears conserved between the two cell types. Murine megakaryocytes have been successfully differentiated in cultures from bone marrow, fetal liver, and embryonic stem (ES) cells, while human megakaryocytes have been cultured from human cord blood, peripheral blood, and ES cells. The various sources of megakaryocyte progenitors provide choices to researchers, allowing them to access the most applicable systems. As examples, both bone marrow-derived and ES cell-derived murine megakaryocytes have been used to study proteins involved in integrin alphaIIbbeta3 regulation such as CIB1 and H-Ras. Therefore, megakaryocytes have provided an invaluable resource for better understanding the biology of platelets. In this chapter, we will describe: (1) approaches to obtain, generate, and characterize megakaryocytes, (2) molecular manipulation of these cells to elevate or decrease expression levels of specific proteins, and (3) current uses and future applications of megakaryocytes.


Asunto(s)
Plaquetas/fisiología , Megacariocitos/fisiología , Animales , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/terapia , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Vectores Genéticos , Humanos , Megacariocitos/citología , Ratones , Interferencia de ARN , Transducción Genética
20.
Circ Res ; 101(11): 1185-93, 2007 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-17975111

RESUMEN

Pathological angiogenesis contributes to various ocular, malignant, and inflammatory disorders, emphasizing the need to understand this process on a molecular level. CIB1 (calcium- and integrin-binding protein), a 22-kDa EF-hand-containing protein, modulates the activity of p21-activated kinase 1 in fibroblasts. Because p21-activated kinase 1 also contributes to endothelial cell function, we hypothesized that CIB1 may have a role in angiogenesis. We found that endothelial cells depleted of CIB1 by either short hairpin RNA or homologous recombination have reduced migration, proliferation, and tubule formation. Moreover, loss of CIB1 in these cells decreases p21-activated kinase 1 activation, downstream extracellular signal-regulated kinase 1/2 activation, and matrix metalloproteinase 2 expression, all of which are known to contribute to angiogenesis. Consistent with these findings, tissues derived from CIB1-deficient (CIB1-/-) mice have reduced growth factor-induced microvessel sprouting in ex vivo organ cultures and in vivo Matrigel plugs. Furthermore, in response to ischemia, CIB1-/- mice demonstrate decreased pathological retinal and adaptive hindlimb angiogenesis. Ischemic CIB1-/- hindlimbs also demonstrate increased tissue damage and significantly reduced p21-activated kinase 1 activation. These data therefore reveal a critical role for CIB1 in ischemia-induced pathological and adaptive angiogenesis.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Células Endoteliales/citología , Isquemia/fisiopatología , Neovascularización Patológica , Neovascularización Fisiológica , Proteínas Angiogénicas/análisis , Animales , Proteínas de Unión al Calcio/deficiencia , Células Endoteliales/fisiología , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Ratones , Ratones Noqueados , Quinasas p21 Activadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA