Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
FASEB J ; 36(7): e22389, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35657599

RESUMEN

The arginine-glycine-aspartic acid (RGD) motif is a cell adhesion sequence that binds to integrins. Some RGD-containing peptides promote adhesion of both embryonic stem cells and induced pluripotent stem cells (iPSCs); however, not all such RGD-containing peptides are active. In this study, we elucidated the role of RGD-neighboring sequences on iPSC adhesion using diverse synthetic peptides and recombinant proteins. Our results indicate that iPSC adhesion requires RGDX1 X2  sequences, such as RGDVF and RGDNY, and that the X1 X2 residues are essential for the adhesion via integrin αvß5 but not αvß3. iPSCs express integrin αvß5 but not αvß3; therefore, iPSC adhesion requires the RGDX1 X2 -containing sequences. The importance of the X1 X2 residues was confirmed with both HeLa and A549 cells, which express integrin αvß5 but not αvß3. Analysis of RGD-neighboring sequences provides important insights into ligand-binding specificity of integrins. Identification of integrin αvß5-binding motifs is potentially useful in drug development, drug delivery, cell culture, and tissue engineering.


Asunto(s)
Células Madre Pluripotentes , Receptores de Vitronectina , Adhesión Celular/fisiología , Integrina alfaVbeta3/genética , Oligopéptidos/química , Péptidos/química , Células Madre Pluripotentes/metabolismo , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo
2.
Sci Rep ; 12(1): 926, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042954

RESUMEN

Lymphangiogenesis is essential for the development of the lymphatic system and is important for physiological processes such as homeostasis, metabolism and immunity. Cellular communication network factor 2 (CCN2, also known as CTGF), is a modular and matricellular protein and a well-known angiogenic factor in physiological and pathological angiogenesis. However, its roles in lymphangiogenesis and intracellular signaling in lymphatic endothelial cells (LECs) remain unclear. Here, we investigated the effects of CCN2 on lymphangiogenesis. In in vivo Matrigel plug assays, exogenous CCN2 increased the number of Podoplanin-positive vessels. Subsequently, we found that CCN2 induced phosphorylation of ERK in primary cultured LECs, which was almost completely inhibited by the blockade of integrin αvß5 and partially decreased by the blockade of integrin αvß3. CCN2 promoted direct binding of ERK to dual-specific phosphatase 6 (DUSP6), which regulated the activation of excess ERK by dephosphorylating ERK. In vitro, CCN2 promoted tube formation in LECs, while suppression of Dusp6 further increased tube formation. In vivo, immunohistochemistry also detected ERK phosphorylation and DUSP6 expression in Podoplanin-positive cells on CCN2-supplemented Matrigel. These results indicated that CCN2 promotes lymphangiogenesis by enhancing integrin αvß5-mediated phosphorylation of ERK and demonstrated that DUSP6 is a negative regulator of excessive lymphangiogenesis by CCN2.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Linfangiogénesis/fisiología , Receptores de Vitronectina/metabolismo , Animales , Movimiento Celular/fisiología , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasa 6 de Especificidad Dual/fisiología , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Femenino , Integrinas/genética , Integrinas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Receptores de Vitronectina/genética , Transducción de Señal/efectos de los fármacos
3.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33903257

RESUMEN

The role of integrins, in particular αv integrins, in regulating insulin resistance is incompletely understood. We have previously shown that the αvß5 integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) regulates cellular uptake of fatty acids. In this work, we evaluated the impact of MFGE8 on glucose homeostasis. We show that acute blockade of the MFGE8/ß5 pathway enhances while acute augmentation dampens insulin-stimulated glucose uptake. Moreover, we find that insulin itself induces cell-surface enrichment of MFGE8 in skeletal muscle, which then promotes interaction between the αvß5 integrin and the insulin receptor leading to dampening of skeletal-muscle insulin receptor signaling. Blockade of the MFGE8/ß5 pathway also enhances hepatic insulin sensitivity. Our work identifies an autoregulatory mechanism by which insulin-stimulated signaling through its cognate receptor is terminated through up-regulation of MFGE8 and its consequent interaction with the αvß5 integrin, thereby establishing a pathway that can potentially be targeted to improve insulin sensitivity.


Asunto(s)
Antígenos de Superficie/genética , Resistencia a la Insulina/genética , Insulina/genética , Proteínas de la Leche/genética , Receptores de Vitronectina/genética , Animales , Antígenos CD/genética , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Glucolípidos/genética , Glicoproteínas/genética , Homeostasis/genética , Humanos , Integrina alfaVbeta3/genética , Gotas Lipídicas , Ratones , Músculo Esquelético/metabolismo , Receptor de Insulina/genética , Transducción de Señal/genética
4.
Sci Rep ; 11(1): 8315, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859325

RESUMEN

The identification of cell surface markers specific to pancreatic beta cells is important for both the study of islet biology and for investigating the pathophysiology of diseases in which this cell type is lost or damaged. Following analysis of publicly available RNAseq data, we identified specific integrin subunits, integrin αv and integrin ß5, that were expressed in beta cells. This finding was further elaborated using immunofluorescence analysis of histological sections derived from donor human pancreas. Despite the broad expression of specific integrin subunits, we found that expression of integrin αvß5 heterodimers was restricted to beta cells and that this complex persisted in islet remnants of some type 1 diabetic individuals from which insulin expression had been lost. This study identifies αvß5 heterodimers as a novel cell surface marker of human pancreatic beta cells, a finding that will aid in the identification and characterisation of this important cell type.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Receptores de Vitronectina/metabolismo , Adulto , Biomarcadores/metabolismo , Femenino , Expresión Génica , Humanos , Insulina/genética , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Vitronectina/genética
5.
FASEB J ; 35(4): e21443, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749877

RESUMEN

The effective clearance of apoptotic cells is an essential step in the resolution of healing wounds. In particular, blood vessel regression during wound resolution produces a significant number of apoptotic endothelial cells (ApoEC) that must be cleared. In considering the fate of ApoEC and the presence of fibroblasts during wound resolution, we hypothesized that fibroblasts might serve as phagocytes involved in endothelial cell removal. The current study investigated whether dermal fibroblasts engulf ApoEC, whether this uptake alters the phenotype of dermal fibroblasts, and the biological molecules involved. In both in vitro and in vivo studies, following ApoEC engulfment, fibroblasts acquired a pro-healing phenotype (increased cell migration, contractility, α-smooth muscle actin expression, and collagen deposition). In addition, fibroblast uptake of ApoEC was shown to be mediated in part by the milk fat globule-EGF factor 8 protein/integrin αv ß5 pathway. Our study demonstrates a novel function of fibroblasts in the clearance of ApoEC and suggests that this capability has significant implications for tissue repair and fibrosis.


Asunto(s)
Células Endoteliales/metabolismo , Piel/irrigación sanguínea , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Apoptosis , Femenino , Proteínas Fluorescentes Verdes , Humanos , Ratones Endogámicos C57BL , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Fagocitosis , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Cicatrización de Heridas
6.
Cell Prolif ; 54(4): e13012, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33656760

RESUMEN

OBJECTIVES: Vitronectin (VTN) has been widely used for the maintenance and expansion of human pluripotent stem cells (hPSCs) as feeder-free conditions. However, the effect of VTN on hPSC differentiation remains unclear. Here, we investigated the role of VTN in early haematopoietic development of hPSCs. MATERIALS AND METHODS: A chemically defined monolayer system was applied to study the role of different matrix or basement membrane proteins in haematopoietic development of hPSCs. The role of integrin signalling in VTN-mediated haematopoietic differentiation was investigated by integrin antagonists. Finally, small interfering RNA was used to knock down integrin gene expression in differentiated cells. RESULTS: We found that the haematopoietic differentiation of hPSCs on VTN was far more efficient than that on Matrigel that is also often used for hPSC culture. VTN promoted the fate determination of endothelial-haematopoietic lineage during mesoderm development to generate haemogenic endothelium (HE). Moreover, we demonstrated that the signals through αvß3 and αvß5 integrins were required for VTN-promoted haematopoietic differentiation. Blocking αvß3 and αvß5 integrins by the integrin antagonists impaired the development of HE, but not endothelial-to-haematopoietic transition (EHT). Finally, both αvß3 and αvß5 were confirmed acting synergistically for early haematopoietic differentiation by knockdown the expression of αv, ß3 or ß5. CONCLUSION: The established VTN-based monolayer system of haematopoietic differentiation of hPSCs presents a valuable platform for further investigating niche signals involved in human haematopoietic development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Integrina alfaVbeta3/metabolismo , Receptores de Vitronectina/metabolismo , Vitronectina/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/genética , Mesodermo/citología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/genética , Transducción de Señal/efectos de los fármacos , Venenos de Serpiente/farmacología
7.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465054

RESUMEN

Group B Streptococcus (GBS) is the major cause of human neonatal infections. A single clone, designated CC17-GBS, accounts for more than 80% of meningitis cases, the most severe form of the infection. However, the events allowing blood-borne GBS to penetrate the brain remain largely elusive. In this study, we identified the host transmembrane receptors α5ß1 and αvß3 integrins as the ligands of Srr2, a major CC17-GBS-specific adhesin. Two motifs located in the binding region of Srr2 were responsible for the interaction between CC17-GBS and these integrins. We demonstrated in a blood-brain-barrier cellular model that both integrins contributed to the adhesion and internalization of CC17-GBS. Strikingly, both integrins were overexpressed during the postnatal period in the brain vessels of the blood-brain barrier and blood-cerebrospinal fluid barrier and contributed to juvenile susceptibility to CC17 meningitis. Finally, blocking these integrins decreased the ability of CC17-GBS to cross into the CNS of juvenile mice in an in vivo model of meningitis. Our study demonstrated that CC17-GBS exploits integrins in order to cross the brain vessels, leading to meningitis. Importantly, it provides host molecular insights into neonate's susceptibility to CC17-GBS meningitis, thereby opening new perspectives for therapeutic and prevention strategies of GBS-elicited meningitis.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Barrera Hematoencefálica/metabolismo , Integrina alfaVbeta3/metabolismo , Meningitis Bacterianas/metabolismo , Receptores de Vitronectina/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus agalactiae/metabolismo , Adhesinas Bacterianas/genética , Animales , Animales Recién Nacidos , Adhesión Bacteriana/genética , Barrera Hematoencefálica/microbiología , Línea Celular , Humanos , Integrina alfaVbeta3/genética , Meningitis Bacterianas/genética , Ratas , Receptores de Vitronectina/genética , Infecciones Estreptocócicas/genética , Streptococcus agalactiae/genética
8.
Methods Mol Biol ; 2217: 3-15, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33215372

RESUMEN

Soluble ligand and conformation-dependent antibody binding assay of integrins on the cell surface is an effective approach to evaluate the activation status of integrins in live cells. The ligands or antibodies are usually labeled with biotin or a fluorescent dye and incubated with integrin-expressing cells in suspension. The cell-bound ligands and antibodies are then detected by flow cytometry. Here we describe the detailed protocols of soluble ligand or antibody binding assay for αIIbß3, αVß3, α5ß1, and αLß2 integrins that are transiently or stably expressed in the model cell lines such as HEK293 or CHO-k1 cells.


Asunto(s)
Bioensayo , Integrina alfaVbeta3/química , Antígeno-1 Asociado a Función de Linfocito/química , Glicoproteína IIb de Membrana Plaquetaria/química , Receptores de Vitronectina/química , Coloración y Etiquetado/métodos , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Células CHO , Adhesión Celular , Cricetulus , Citometría de Flujo , Colorantes Fluorescentes/química , Células HEK293 , Humanos , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Molécula 1 de Adhesión Intercelular , Ligandos , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Plásmidos/química , Plásmidos/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Unión Proteica , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
9.
J Cell Biol ; 219(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33141174

RESUMEN

Fibronectin (FN) is an essential glycoprotein of the extracellular matrix; binds integrins, syndecans, collagens, and growth factors; and is assembled by cells into complex fibrillar networks. The RGD motif in FN facilitates cell binding- and fibrillogenesis through binding to α5ß1 and αv-class integrins. However, whether RGD is the sole binding site for αv-class integrins is unclear. Most notably, substituting aspartate with glutamate (RGE) was shown to eliminate integrin binding in vitro, while mouse genetics revealed that FNRGE preserves αv-class integrin binding and fibrillogenesis. To address this conflict, we employed single-cell force spectroscopy, engineered cells, and RGD motif-deficient mice (Fn1ΔRGD/ΔRGD) to search for additional αv-class integrin-binding sites. Our results demonstrate that α5ß1 and αv-class integrins solely recognize the FN-RGD motif and that αv-class, but not α5ß1, integrins retain FN-RGE binding. Furthermore, Fn1ΔRGD/ΔRGD tissues and cells assemble abnormal and dysfunctional FNΔRGD fibrils in a syndecan-dependent manner. Our data highlight the central role of FN-RGD and the functionality of FN-RGE for αv-class integrins.


Asunto(s)
Mutación , Oligopéptidos/metabolismo , Animales , Ratones , Ratones Mutantes , Oligopéptidos/genética , Receptores de Vitronectina/genética
10.
mBio ; 11(3)2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32487760

RESUMEN

Mucormycosis, caused by Rhizopus species, is a life-threatening fungal infection that occurs in patients immunocompromised by diabetic ketoacidosis (DKA), cytotoxic chemotherapy, immunosuppressive therapy, hematologic malignancies, or severe trauma. Inhaled Rhizopus spores cause pulmonary infections in patients with hematologic malignancies, while patients with DKA are much more prone to rhinoorbital/cerebral mucormycosis. Here, we show that Rhizopus delemar interacts with glucose-regulated protein 78 (GRP78) on nasal epithelial cells via its spore coat protein CotH3 to invade and damage the nasal epithelial cells. Expression of the two proteins is significantly enhanced by high glucose, iron, and ketone body levels (hallmark features of DKA), potentially leading to frequently lethal rhinoorbital/cerebral mucormycosis. In contrast, R. delemar CotH7 recognizes integrin ß1 as a receptor on alveolar epithelial cells, causing the activation of epidermal growth factor receptor (EGFR) and leading to host cell invasion. Anti-integrin ß1 antibodies inhibit R. delemar invasion of alveolar epithelial cells and protect mice from pulmonary mucormycosis. Our results show that R. delemar interacts with different mammalian receptors depending on the host cell type. Susceptibility of patients with DKA primarily to rhinoorbital/cerebral disease can be explained by host factors typically present in DKA and known to upregulate CotH3 and nasal GRP78, thereby trapping the fungal cells within the rhinoorbital milieu, leading to subsequent invasion and damage. Our studies highlight that mucormycosis pathogenesis can potentially be overcome by the development of novel customized therapies targeting niche-specific host receptors or their respective fungal ligands.IMPORTANCE Mucormycosis caused by Rhizopus species is a fungal infection with often fatal prognosis. Inhalation of spores is the major route of entry, with nasal and alveolar epithelial cells among the first cells that encounter the fungi. In patients with hematologic malignancies or those undergoing cytotoxic chemotherapy, Rhizopus causes pulmonary infections. On the other hand, DKA patients predominantly suffer from rhinoorbital/cerebral mucormycosis. The reason for such disparity in disease types by the same fungus is not known. Here, we show that the unique susceptibility of DKA subjects to rhinoorbital/cerebral mucormycosis is likely due to specific interaction between nasal epithelial cell GRP78 and fungal CotH3, the expression of which increases in the presence of host factors present in DKA. In contrast, pulmonary mucormycosis is initiated via interaction of inhaled spores expressing CotH7 with integrin ß1 receptor, which activates EGFR to induce fungal invasion of host cells. These results introduce a plausible explanation for disparate disease manifestations in DKA versus those in hematologic malignancy patients and provide a foundation for development of therapeutic interventions against these lethal forms of mucormycosis.


Asunto(s)
Células Epiteliales/microbiología , Proteínas de Choque Térmico/genética , Interacciones Huésped-Patógeno , Infecciones Fúngicas Invasoras/microbiología , Mucormicosis/microbiología , Receptores de Vitronectina/genética , Rhizopus/patogenicidad , Células A549 , Células Epiteliales Alveolares/microbiología , Células Epiteliales Alveolares/patología , Animales , Línea Celular , Cetoacidosis Diabética/complicaciones , Cetoacidosis Diabética/microbiología , Chaperón BiP del Retículo Endoplásmico , Células Epiteliales/patología , Receptores ErbB/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Nariz/citología , Virulencia
11.
Cell Rep ; 30(4): 969-983.e4, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31956073

RESUMEN

We perform a CRISPR-Cas9 genome-wide screen in glioblastoma stem cells and identify integrin αvß5 as an internalization factor for Zika virus (ZIKV). Expression of αvß5 is correlated with ZIKV susceptibility in various cells and tropism in developing human cerebral cortex. A blocking antibody against integrin αvß5, but not αvß3, efficiently inhibits ZIKV infection. ZIKV binds to cells but fails to internalize when treated with integrin αvß5-blocking antibody. αvß5 directly binds to ZIKV virions and activates focal adhesion kinase, which is required for ZIKV infection. Finally, αvß5 blocking antibody or two inhibitors, SB273005 and cilengitide, reduces ZIKV infection and alleviates ZIKV-induced pathology in human neural stem cells and in mouse brain. Altogether, our findings identify integrin αvß5 as an internalization factor for ZIKV, providing a promising therapeutic target, as well as two drug candidates for prophylactic use or treatments for ZIKV infections.


Asunto(s)
Antivirales/uso terapéutico , Células-Madre Neurales/metabolismo , Receptores de Vitronectina/genética , Infección por el Virus Zika/virología , Virus Zika/patogenicidad , Antivirales/farmacología , Humanos , Receptores de Vitronectina/metabolismo
12.
Bioconjug Chem ; 30(11): 2777-2781, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31657906

RESUMEN

Learning from the design concept of antibody-drug conjugates (ADCs), we attempted to construct siRNA conjugated polymer brush by attaching a multiple of siRNA to the units of poly(amino acids) [poly(lysine) derivatives] through an intracellular cleavable disulfide bond. Note that the disulfide linkage is stable at extracellular milieu yet subjected to cleavage into free thiol residues at the intracellular reducing compartments. Consequently, ready release of arrays of active siRNA was achieved selectively in the intracellular compartments. Furthermore, tumor-targeted cyclic Asp-Gly-Arg (RGD) was conjugated to the aforementioned polymer brush in view that the RGD receptors (αVß3 and αVß5 integrins) were overexpressed over a wide spectrum of cancerous cells. Our subsequent results have achieved potent gene silencing in cultured cancerous cells from our proposed siRNA delivery construct. To our best knowledge, our proposed conjugate should be the first example of using an ADC platform in successful intracellular transportation of larger macromolecular biological payloads rather than small molecular chemotherapeutic drugs. Hence, the proposed strategy may serve as a promising avenue for targeted delivery of macromolecular pharmaceutical payloads.


Asunto(s)
Silenciador del Gen , Glioma/genética , Integrina alfaVbeta3/antagonistas & inhibidores , Oligopéptidos/química , Polímeros/química , ARN Interferente Pequeño/administración & dosificación , Receptores de Vitronectina/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Glioma/metabolismo , Glioma/patología , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Oxidación-Reducción , ARN Interferente Pequeño/genética , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Células Tumorales Cultivadas
13.
J Cell Sci ; 132(20)2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31515275

RESUMEN

Diurnal clearance phagocytosis by the retinal pigment epithelium (RPE) is a conserved efferocytosis process whose binding step is mediated by αvß5 integrin receptors. Two related annexins, A5 (ANXA5) and A6 (ANXA6), share an αvß5 integrin-binding motif. Here, we report that ANXA5, but not ANXA6, regulates the binding capacity for spent photoreceptor outer segment fragments or apoptotic cells by fibroblasts and RPE. Similar to αvß5-deficient RPE, ANXA5-/- RPE in vivo lacks the diurnal burst of phagocytosis that follows photoreceptor shedding in wild-type retina. Increasing ANXA5 in cells lacking αvß5 or increasing αvß5 in cells lacking ANXA5 does not affect particle binding. Association of cytosolic ANXA5 and αvß5 integrin in RPE in culture and in vivo further supports their functional interdependence. Silencing ANXA5 is sufficient to reduce levels of αvß5 receptors at the apical phagocytic surface of RPE cells. The effect of ANXA5 on surface αvß5 and on particle binding requires the C-terminal ANXA5 annexin repeat but not its unique N-terminus. These results identify a novel role for ANXA5 specifically in the recognition and binding step of clearance phagocytosis, which is essential to retinal physiology.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Anexina A5/metabolismo , Apoptosis , Fagocitosis , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Vitronectina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Anexina A5/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Células Fotorreceptoras de Vertebrados/citología , Ratas , Ratas Sprague-Dawley , Receptores de Vitronectina/genética , Epitelio Pigmentado de la Retina/citología
14.
Structure ; 27(9): 1443-1451.e6, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31353240

RESUMEN

Targeting both integrins αVß3 and α5ß1 simultaneously appears to be more effective in cancer therapy than targeting each one alone. The structural requirements for bispecific binding of ligand to integrins have not been fully elucidated. RGD-containing knottin 2.5F binds selectively to αVß3 and α5ß1, whereas knottin 2.5D is αVß3 specific. To elucidate the structural basis of this selectivity, we determined the structures of 2.5F and 2.5D as apo proteins and in complex with αVß3, and compared their interactions with integrins using molecular dynamics simulations. These studies show that 2.5D engages αVß3 by an induced fit, but conformational selection of a flexible RGD loop accounts for high-affinity selective binding of 2.5F to both integrins. The contrasting binding of the highly flexible low-affinity linear RGD peptides to multiple integrins suggests that a "Goldilocks zone" of conformational flexibility of the RGD loop in 2.5F underlies its selective binding promiscuity to integrins.


Asunto(s)
Miniproteínas Nodales de Cistina/metabolismo , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Receptores de Vitronectina/química , Receptores de Vitronectina/metabolismo , Sitios de Unión , Humanos , Integrina alfaVbeta3/genética , Células K562 , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica , Receptores de Vitronectina/genética
15.
J Clin Invest ; 129(1): 137-149, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30307407

RESUMEN

Glioblastoma is highly enriched with macrophages, and osteopontin (OPN) expression levels correlate with glioma grade and the degree of macrophage infiltration; thus, we studied whether OPN plays a crucial role in immune modulation. Quantitative PCR, immunoblotting, and ELISA were used to determine OPN expression. Knockdown of OPN was achieved using complementary siRNA, shRNA, and CRISPR/Cas9 techniques, followed by a series of in vitro functional migration and immunological assays. OPN gene-deficient mice were used to examine the roles of non-tumor-derived OPN on survival of mice harboring intracranial gliomas. Patients with mesenchymal glioblastoma multiforme (GBM) show high OPN expression, a negative survival prognosticator. OPN is a potent chemokine for macrophages, and its blockade significantly impaired the ability of glioma cells to recruit macrophages. Integrin αvß5 (ITGαvß5) is highly expressed on glioblastoma-infiltrating macrophages and constitutes a major OPN receptor. OPN maintains the M2 macrophage gene signature and phenotype. Both tumor-derived and host-derived OPN were critical for glioma development. OPN deficiency in either innate immune or glioma cells resulted in a marked reduction in M2 macrophages and elevated T cell effector activity infiltrating the glioma. Furthermore, OPN deficiency in the glioma cells sensitized them to direct CD8+ T cell cytotoxicity. Systemic administration in mice of 4-1BB-OPN bispecific aptamers was efficacious, increasing median survival time by 68% (P < 0.05). OPN is thus an important chemokine for recruiting macrophages to glioblastoma, mediates crosstalk between tumor cells and the innate immune system, and has the potential to be exploited as a therapeutic target.


Asunto(s)
Neoplasias Encefálicas/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Glioblastoma/inmunología , Inmunidad Innata , Macrófagos/inmunología , Proteínas de Neoplasias/inmunología , Osteopontina/inmunología , Animales , Aptámeros de Nucleótidos/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Osteopontina/genética , Receptores de Vitronectina/genética , Receptores de Vitronectina/inmunología
16.
Oncogene ; 38(8): 1282-1295, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30254208

RESUMEN

Phenotypic plasticity and subsequent generation of intratumoral heterogeneity underly key traits in malignant melanoma such as drug resistance and metastasis. Melanoma plasticity promotes a switch between proliferative and invasive phenotypes characterized by different transcriptional programs of which MITF is a critical regulator. Here, we show that the acid ceramidase ASAH1, which controls sphingolipid metabolism, acted as a rheostat of the phenotypic switch in melanoma cells. Low ASAH1 expression was associated with an invasive behavior mediated by activation of the integrin alphavbeta5-FAK signaling cascade. In line with that, human melanoma biopsies revealed heterogeneous staining of ASAH1 and low ASAH1 expression at the melanoma invasive front. We also identified ASAH1 as a new target of MITF, thereby involving MITF in the regulation of sphingolipid metabolism. Together, our findings provide new cues to the mechanisms underlying the phenotypic plasticity of melanoma cells and identify new anti-metastatic targets.


Asunto(s)
Ceramidasa Ácida/genética , Proliferación Celular/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Línea Celular Tumoral , Femenino , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/patología , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas B-raf , Receptores de Vitronectina/genética , Transducción de Señal
17.
J Transl Med ; 16(1): 352, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30541573

RESUMEN

BACKGROUND: To date the TGF-ß1 activation mediated by integrin ανß5 during fibrosis is well-known. This process has been shown also in the heart, where cardiac fibroblasts (CF) differentiate into α-smooth muscle actin (α-SMA)-positive myofibroblasts (MyoFB). Here, we studied the effects on CF, isolated by spontaneously hypertensive rats (SHR), of integrin ανß5 inhibition in MyoFB differentiation. METHODS: Staining and immunohistochemistry were performed on rat cardiac tissue. CF were isolated by enzymatic digestion from SHR (SHR-CF) and normotensive WKY (WKY-CF) rat hearts and then treated for in vitro evaluation. RESULTS: SHR heart tissues revealed a higher TGF-ß1 expression vs. WKY samples. SHR-CF showed an enhanced SMAD2/3 activation and an up-regulated expression of α-SMA, a typical MyoFB marker, especially after TGF-ß1 treatment. Immunostaining on cardiac tissues revealed a higher expression of integrin ανß5 in SHR vs. WKY rat hearts. In vitro results confirmed the up-regulation of integrin ανß5 expression in SHR-CF at basal condition and after TGF-ß1 treatment, in comparison with WKY-CF. Inhibition of integrin ανß5 by cilengitide treatment led a decreased expression of ανß5, collagen I, and α-SMA in SHR-CF vs. WKY-CF, resulting in a diminished differentiation of CF into MyoFB. Taking together, results suggested that SHR-CF are more susceptible to TGF-ß1, showing an up-regulated activation of SMAD2/3 signaling, and an increased ανß5, α-SMA, and collagen I expression. Hypertension stimulus promoted an up-regulation of integrin ανß5 on SHR cardiac tissue and its in vitro inhibition reverted pro-fibrotic events of SHR-CF. CONCLUSION: Inhibition of integrin ανß5 exerted by cilengitide strongly diminished SHR-CF differentiation into detrimental MyoFB. So, integrin ανß5 might be considered a novel therapeutic target and cilengitide an effective pharmacological tool to limit the progression of hypertension-induced cardiac fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Miocardio/metabolismo , Miocardio/patología , Receptores de Vitronectina/antagonistas & inhibidores , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Presión Sanguínea/efectos de los fármacos , Colágeno Tipo I/metabolismo , Diástole/efectos de los fármacos , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Venenos de Serpiente/farmacología , Sístole/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
18.
Int J Oncol ; 53(6): 2683-2694, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30280197

RESUMEN

The malignancy of glioblastoma (GB) is primarily due to the ability of glioma cancer stem cells (GSC) to disseminate into surrounding brain tissues, despite surgery and chemotherapy, and to form new tumoral masses. Members of the RGD-binding integrin family, which recognize the arginine-glycine-aspartic acid (RGD) sequence present in components of the extracellular matrix, and which serve a crucial function in the dissemination of GCS, are overexpressed in GB. Small-molecule integrin antagonists (SMIAs) designed to recognize RGD-integrins may therefore be an effective tool for decreasing GB infiltration and recurrence. In the present study, in vitro pro-apoptotic and infiltrative effects elicited by the SMIA 1a­RGD in human GSC were investigated. Reverse transcription-quantitative polymerase chain reaction analysis revealed that, compared with normal human astrocytes, GSC grown on laminin-coated dishes overexpressed stemness markers as well as αvß3 and αvß5 integrins. In addition, dissociated GSC were identified to exhibit tumorigenic capacity when injected into immunodeficient mice. Using annexin/fluorescence-activated cell sorting analysis and ELISA nucleosome assays, it was identified that treatment of GSC with 25 µM 1a­RGD for 48 h elicited detachment­dependent anoikis not accompanied by necrosis-dependent cell death. A colorimetric proliferation assay indicated that 1a­RGD did not affect cell viability, but that, instead, it markedly inhibited GSC migration as assessed using a Transwell assay. Western blot experiments revealed a decrease in focal adhesion kinase and protein kinase B phosphorylation with a concomitant increase in caspase-9 and -3/7 activity following 1a­RGD treatment, suggesting that the pro-anoikis effects of 1a­RGD may be mediated by these molecular mechanisms. Western blot analysis revealed no changes in specific markers of autophagy, suggesting further that 1a­RGD-induced cell death is primarily sustained by anoikis-associated mechanisms. In conclusion, the results of the present study indicate that SMIA have potential as a therapeutic tool for decreasing GSC dissemination.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Integrinas/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Anciano , Animales , Anoicis , Neoplasias Encefálicas/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/genética , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/genética , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacología
19.
J Clin Invest ; 128(11): 5018-5033, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295643

RESUMEN

Emerging evidence indicates that angiopoietin-2 (Angpt2), a well-recognized vascular destabilizing factor, is a biomarker of poor outcome in ischemic heart disease. However, its precise role in postischemic cardiovascular remodeling is poorly understood. Here, we show that Angpt2 plays multifaceted roles in the exacerbation of cardiac hypoxia and inflammation after myocardial ischemia. Angpt2 was highly expressed in endothelial cells at the infarct border zone after myocardial infarction (MI) or ischemia/reperfusion injury in mice. In the acute phase of MI, endothelial-derived Angpt2 antagonized Angpt1/Tie2 signaling, which was greatly involved in pericyte detachment, vascular leakage, increased adhesion molecular expression, degradation of the glycocalyx and extracellular matrix, and enhanced neutrophil infiltration and hypoxia in the infarct border area. In the chronic remodeling phase after MI, endothelial- and macrophage-derived Angpt2 continuously promoted abnormal vascular remodeling and proinflammatory macrophage polarization through integrin α5ß1 signaling, worsening cardiac hypoxia and inflammation. Accordingly, inhibition of Angpt2 either by gene deletion or using an anti-Angpt2 blocking antibody substantially alleviated these pathological findings and ameliorated postischemic cardiovascular remodeling. Blockade of Angpt2 thus has potential as a therapeutic option for ischemic heart failure.


Asunto(s)
Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Transducción de Señal , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/genética , Animales , Anticuerpos Bloqueadores/farmacología , Células Endoteliales/patología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética
20.
BMC Biol ; 16(1): 92, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30119679

RESUMEN

BACKGROUND: Increased activity of the receptor tyrosine kinase Tie2 has been implicated in the promotion of pathological angiogenesis. This activity is mainly mediated through angiopoietin (Ang)1- and Ang2-dependent activation of integrins by Tie2, rendering the Ang/Tie2/integrin axis an attractive putative target for cancer therapeutics. RESULTS: To target this axis, we developed single domain, non-immunoglobulin high-affinity bi-specific protein inhibitors against both Tie2 and αvß3 integrin. We have previously engineered the Ang2-binding domain of Tie2 (Ang2-BD) as a Tie2 inhibitor. Here, we engineered an exposed loop in Ang2-BD to generate variants that include an integrin-binding Arg-Gly-Asp (RGD) motif and used flow cytometry screening of a yeast-displayed Ang2-BD RGD loop library to identify the integrin antagonists. The bi-specific antagonists targeting both Tie2 and αvß3 integrin inhibited adhesion and proliferation of endothelial cells cultured together with the αvß3 integrin ligand vitronectin, as well as endothelial cell invasion and tube formation. The bi-specific reagents inhibited downstream signaling by Tie2 intracellularly in response to its agonist Ang1 more effectively than the wild-type Ang2 BD that binds Tie2 alone. CONCLUSIONS: Collectively, this study-the first to describe inhibitors targeting all the known functions resulting from Tie2/integrin αvß3 cross-talk-has created new tools for studying Tie2- and integrin αvß3-dependent molecular pathways and provides the basis for the rational and combinatorial engineering of ligand-Tie2 and ligand-integrin αvß3 receptor interactions. Given the roles of these pathways in cancer angiogenesis and metastasis, this proof of principle study paves the route to create novel Tie2/integrin αvß3-targeting proteins for clinical use as imaging and therapeutic agents.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Fisiológica/genética , Receptor TIE-2/antagonistas & inhibidores , Receptores de Vitronectina/genética , Ribonucleasa Pancreática/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Animales , Ratones , Receptor TIE-2/química , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Receptores de Vitronectina/química , Receptores de Vitronectina/metabolismo , Ribonucleasa Pancreática/química , Ribonucleasa Pancreática/genética , Ribonucleasa Pancreática/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...