Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 24(6): 1007-1019, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37069398

RESUMO

Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR-Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 ß-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of ßII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1-ßII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell immunotherapy.


Assuntos
Receptores de Antígenos Quiméricos , Animais , Camundongos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Movimento Celular , Imunoterapia Adotiva , Antígeno-1 Associado à Função Linfocitária , Espectrina , Humanos , Feminino
2.
Front Immunol ; 12: 666231, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149701

RESUMO

Although cancer immunotherapy is effective against hematological malignancies, it is less effective against solid tumors due in part to significant metabolic challenges present in the tumor microenvironment (TME), where infiltrated CD8+ T cells face fierce competition with cancer cells for limited nutrients. Strong metabolic suppression in the TME is often associated with impaired T cell recruitment to the tumor site and hyporesponsive effector function via T cell exhaustion. Increasing evidence suggests that mitochondria play a key role in CD8+ T cell activation, effector function, and persistence in tumors. In this study, we showed that there was an increase in overall mitochondrial function, including mitochondrial mass and membrane potential, during both mouse and human CD8+ T cell activation. CD8+ T cell mitochondrial membrane potential was closely correlated with granzyme B and IFN-γ production, demonstrating the significance of mitochondria in effector T cell function. Additionally, activated CD8+ T cells that migrate on ICAM-1 and CXCL12 consumed significantly more oxygen than stationary CD8+ T cells. Inhibition of mitochondrial respiration decreased the velocity of CD8+ T cell migration, indicating the importance of mitochondrial metabolism in CD8+ T cell migration. Remote optical stimulation of CD8+ T cells that express our newly developed "OptoMito-On" successfully enhanced mitochondrial ATP production and improved overall CD8+ T cell migration and effector function. Our study provides new insight into the effect of the mitochondrial membrane potential on CD8+ T cell effector function and demonstrates the development of a novel optogenetic technique to remotely control T cell metabolism and effector function at the target tumor site with outstanding specificity and temporospatial resolution.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos da radiação , Optogenética/métodos , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/efeitos da radiação , Citocinas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/efeitos da radiação , Humanos , Imunoterapia , Ativação Linfocitária/efeitos da radiação , Potencial da Membrana Mitocondrial/genética , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Neoplasias/imunologia , Neoplasias/terapia
3.
Integr Biol (Camb) ; 12(11): 275-289, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33164044

RESUMO

Endothelial cells (ECs) are an active component of the immune system and interact directly with inflammatory cytokines. While ECs are known to be polarized cells, the potential role of apicobasal polarity in response to inflammatory mediators has been scarcely studied. Acute inflammation is vital in maintaining healthy tissue in response to infection; however, chronic inflammation can lead to the production of systemic inflammatory cytokines and deregulated leukocyte trafficking, even in the absence of a local infection. Elevated levels of cytokines in circulation underlie the pathogenesis of sepsis, the leading cause of intensive care death. Because ECs constitute a key barrier between circulation (luminal interface) and tissue (abluminal interface), we hypothesize that ECs respond differentially to inflammatory challenge originating in the tissue versus circulation as in local and systemic inflammation, respectively. To begin this investigation, we stimulated ECs abluminally and luminally with the inflammatory cytokine tumor necrosis factor alpha (TNF-α) to mimic a key feature of local and systemic inflammation, respectively, in a microvascular mimetic (µSiM-MVM). Polarized IL-8 secretion and polymorphonuclear neutrophil (PMN) transmigration were quantified to characterize the EC response to luminal versus abluminal TNF-α. We observed that ECs uniformly secrete IL-8 in response to abluminal TNF-α and is followed by PMN transmigration. The response to abluminal treatment was coupled with the formation of ICAM-1-rich membrane ruffles on the apical surface of ECs. In contrast, luminally stimulated ECs secreted five times more IL-8 into the luminal compartment than the abluminal compartment and sequestered PMNs on the apical EC surface. Our results identify clear differences in the response of ECs to TNF-α originating from the abluminal versus luminal side of a monolayer for the first time and may provide novel insight into future inflammatory disease intervention strategies.


Assuntos
Biomimética , Sistema Imunitário , Microcirculação , Fator de Necrose Tumoral alfa/metabolismo , Adesão Celular , Comunicação Celular/fisiologia , Movimento Celular , Citocinas/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Inflamação , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-8/metabolismo , Microfluídica , Microscopia de Fluorescência , Neutrófilos/citologia , Permeabilidade , Sepse/microbiologia
4.
Small ; 15(6): e1804111, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30632319

RESUMO

Selective cellular transmigration across the microvascular endothelium regulates innate and adaptive immune responses, stem cell localization, and cancer cell metastasis. Integration of traditional microporous membranes into microfluidic vascular models permits the rapid assay of transmigration events but suffers from poor reproduction of the cell permeable basement membrane. Current microporous membranes in these systems have large nonporous regions between micropores that inhibit cell communication and nutrient exchange on the basolateral surface reducing their physiological relevance. Here, the use of 100 nm thick continuously nanoporous silicon nitride membranes as a base substrate for lithographic fabrication of 3 µm pores is presented, resulting in a highly porous (≈30%), dual-scale nano- and microporous membrane for use in an improved vascular transmigration model. Ultrathin membranes are patterned using a precision laser writer for cost-effective, rapid micropore design iterations. The optically transparent dual-scale membranes enable complete observation of leukocyte egress across a variety of pore densities. A maximal density of ≈14 micropores per cell is discovered beyond which cell-substrate interactions are compromised giving rise to endothelial cell losses under flow. Addition of a subluminal extracellular matrix rescues cell adhesion, allowing for the creation of shear-primed endothelial barrier models on nearly 30% continuously porous substrates.


Assuntos
Células Endoteliais da Veia Umbilical Humana/citologia , Membranas Artificiais , Modelos Biológicos , Nanopartículas/química , Migração Transendotelial e Transepitelial , Animais , Adesão Celular , Colágeno/metabolismo , Matriz Extracelular/química , Géis/química , Humanos , Nanopartículas/ultraestrutura , Nanoporos/ultraestrutura , Neutrófilos/citologia , Porosidade , Ratos
5.
Biomed Microdevices ; 20(1): 11, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29305767

RESUMO

Silicon nanomembranes are ultrathin, highly permeable, optically transparent and biocompatible substrates for the construction of barrier tissue models. Trans-epithelial/endothelial electrical resistance (TEER) is often used as a non-invasive, sensitive and quantitative technique to assess barrier function. The current study characterizes the electrical behavior of devices featuring silicon nanomembranes to facilitate their application in TEER studies. In conventional practice with commercial systems, raw resistance values are multiplied by the area of the membrane supporting cell growth to normalize TEER measurements. We demonstrate that under most circumstances, this multiplication does not 'normalize' TEER values as is assumed, and that the assumption is worse if applied to nanomembrane chips with a limited active area. To compare the TEER values from nanomembrane devices to those obtained from conventional polymer track-etched (TE) membranes, we develop finite element models (FEM) of the electrical behavior of the two membrane systems. Using FEM and parallel cell-culture experiments on both types of membranes, we successfully model the evolution of resistance values during the growth of endothelial monolayers. Further, by exploring the relationship between the models we develop a 'correction' function, which when applied to nanomembrane TEER, maps to experiments on conventional TE membranes. In summary, our work advances the the utility of silicon nanomembranes as substrates for barrier tissue models by developing an interpretation of TEER values compatible with conventional systems.


Assuntos
Impedância Elétrica , Análise de Elementos Finitos , Membranas Artificiais , Nanoestruturas/química , Animais , Barreira Hematoencefálica , Encéfalo/citologia , Células Cultivadas , Eletrodos , Endotélio Vascular/citologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Camundongos , Modelos Teóricos , Permeabilidade , Reprodutibilidade dos Testes , Silício
6.
Sci Rep ; 7(1): 5164, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701737

RESUMO

Hematopoietic ontogeny is characterized by distinct primitive and definitive erythroid lineages. Definitive erythroblasts mature and enucleate extravascularly and form a unique membrane skeleton, composed of spectrin, 4.1R-complex, and ankyrinR-complex components, to survive the vicissitudes of the adult circulation. However, little is known about the formation and composition of the membrane skeleton in primitive erythroblasts, which progressively mature while circulating in the embryonic bloodstream. We found that primary primitive erythroblasts express the major membrane skeleton genes present in similarly staged definitive erythroblasts, suggesting that the composition and formation of this membrane network is conserved in maturing primitive and definitive erythroblasts despite their respective intravascular and extravascular locations. Membrane deformability and stability of primitive erythroblasts, assayed by microfluidic studies and fluorescence imaged microdeformation, respectively, significantly increase prior to enucleation. These functional changes coincide with protein 4.1 R isoform switching and protein 4.1R-null primitive erythroblasts fail to establish normal membrane stability and deformability. We conclude that maturing primitive erythroblasts initially navigate the embryonic vasculature prior to establishing a deformable cytoskeleton, which is ultimately formed prior to enucleation. Formation of an erythroid-specific, protein 4.1R-dependent membrane skeleton is an important feature not only of definitive, but also of primitive, erythropoiesis in mammals.


Assuntos
Diferenciação Celular , Eritroblastos/metabolismo , Eritropoese , Proteínas dos Microfilamentos/metabolismo , Processamento Alternativo , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Eritroblastos/citologia , Membrana Eritrocítica/metabolismo , Eritropoese/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética
7.
J Biomed Mater Res A ; 105(4): 1112-1122, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28093865

RESUMO

Poly(ethylene glycol) (PEG) hydrogels provide a versatile platform to develop cell instructive materials through incorporation of a variety of cell adhesive ligands and degradable chemistries. Synthesis of PEG gels can be accomplished via two mechanisms: chain and step growth polymerizations. The mechanism dramatically impacts hydrogel nanostructure, whereby chain polymerized hydrogels are highly heterogeneous and step growth networks exhibit more uniform structures. Underpinning these alterations in nanostructure of chain polymerized hydrogels are densely-packed hydrophobic poly(methyl methacrylate) or poly(acrylate) kinetic chains between hydrophilic PEG crosslinkers. As cell-material interactions, such as those mediated by integrins, occur at the nanoscale and affect cell behavior, it is important to understand how different modes of polymerization translate into nanoscale mechanical and hydrophobic heterogeneities of hydrogels. Therefore, chain- and step-growth polymerized PEG hydrogels with macroscopically similar macromers and compliance (for example, methacrylate-functionalized PEG (PEGDM), MW = 10 kDa and norbornene-functionalized 4-arm PEG (PEGnorb), MW = 10 kDa) were used to examine potential nanoscale differences in hydrogel mechanics and hydrophobicity using atomic force microscopy (AFM). It was found that chain-growth polymerized network yielded greater heterogeneities in both stiffness and hydrophobicity as compared to step-growth polymerized networks. These nanoscale heterogeneities impact cell-material interactions, particularly human mesenchymal stem cell (hMSC) adhesion and spreading, which has implications in use of these hydrogels for tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1112-1122, 2017.


Assuntos
Resinas Acrílicas , Hidrogéis , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Polietilenoglicóis , Polimetil Metacrilato , Resinas Acrílicas/química , Resinas Acrílicas/farmacologia , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polimetil Metacrilato/química , Polimetil Metacrilato/farmacologia
8.
Langmuir ; 31(50): 13553-60, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26605493

RESUMO

The nanoscale topography of adhesive surfaces is known to be an important factor governing cellular behavior. Previous work has shown that surface coatings composed of halloysite nanotubes enhance the adhesion, and therefore capture of, rare target cells such as circulating tumor cells. Here we demonstrate a unique feature of these coatings in their ability to reduce the adhesion of leukocytes and prevent leukocyte spreading. Surfaces were prepared with coatings of halloysite nanotubes and functionalized for leukocyte adhesion with E-selectin, and the dilution of nanotube concentration revealed a threshold concentration below which cell spreading became comparable to smooth surfaces. Evaluation of surface roughness characteristics determined that the average distance between discrete surface features correlated with adhesion metrics, with a separation distance of ∼2 µm identified as the critical threshold. Computational modeling of the interaction of leukocytes with halloysite nanotube-coated surfaces of varying concentrations demonstrates that the geometry of the cell surface and adhesive counter-surface produces a significantly diminished effective contact area compared to a leukocyte interacting with a smooth surface.


Assuntos
Forma Celular , Leucócitos/citologia , Nanotubos/química , Adesão Celular , Voluntários Saudáveis , Humanos , Tamanho da Partícula , Porosidade , Propriedades de Superfície
9.
Proc Natl Acad Sci U S A ; 112(38): 11783-8, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26351678

RESUMO

Piezo proteins (Piezo1 and Piezo2) are recently identified mechanically activated cation channels in eukaryotic cells and associated with physiological responses to touch, pressure, and stretch. In particular, human RBCs express Piezo1 on their membranes, and mutations of Piezo1 have been linked to hereditary xerocytosis. To date, however, physiological functions of Piezo1 on normal RBCs remain poorly understood. Here, we show that Piezo1 regulates mechanotransductive release of ATP from human RBCs by controlling the shear-induced calcium (Ca(2+)) influx. We find that, in human RBCs treated with Piezo1 inhibitors or having mutant Piezo1 channels, the amounts of shear-induced ATP release and Ca(2+) influx decrease significantly. Remarkably, a critical extracellular Ca(2+) concentration is required to trigger significant ATP release, but membrane-associated ATP pools in RBCs also contribute to the release of ATP. Our results show how Piezo1 channels are likely to function in normal RBCs and suggest a previously unidentified mechanotransductive pathway in ATP release. Thus, we anticipate that the study will impact broadly on the research of red cells, cellular mechanosensing, and clinical studies related to red cell disorders and vascular disease.


Assuntos
Trifosfato de Adenosina/metabolismo , Eritrócitos/metabolismo , Canais Iônicos/metabolismo , Mecanotransdução Celular , Cálcio/metabolismo , Calibragem , Membrana Eritrocítica/metabolismo , Espaço Extracelular/metabolismo , Humanos , Microfluídica , Modelos Biológicos , Resistência ao Cisalhamento
10.
Stem Cell Reports ; 4(6): 995-1003, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26028528

RESUMO

Red blood cells (RBCs), responsible for oxygen delivery and carbon dioxide exchange, are essential for our well-being. Alternative RBC sources are needed to meet the increased demand for RBC transfusions projected to occur as our population ages. We previously have discovered that erythroblasts derived from the early mouse embryo can self-renew extensively ex vivo for many months. To better understand the mechanisms regulating extensive erythroid self-renewal, global gene expression data sets from self-renewing and differentiating erythroblasts were analyzed and revealed the differential expression of Bmi-1. Bmi-1 overexpression conferred extensive self-renewal capacity upon adult bone-marrow-derived self-renewing erythroblasts, which normally have limited proliferative potential. Importantly, Bmi-1 transduction did not interfere with the ability of extensively self-renewing erythroblasts (ESREs) to terminally mature either in vitro or in vivo. Bmi-1-induced ESREs can serve to generate in vitro models of erythroid-intrinsic disorders and ultimately may serve as a source of cultured RBCs for transfusion therapy.


Assuntos
Eritroblastos/citologia , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Dexametasona/farmacologia , Eritroblastos/metabolismo , Eritroblastos/transplante , Eritropoetina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Células-Tronco/farmacologia , Irradiação Corporal Total
11.
Proc Natl Acad Sci U S A ; 109(26): 10474-9, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22689994

RESUMO

T cells spend the majority of their time perusing lymphoid organs in search of cognate antigen presented by antigen presenting cells (APCs) and then quickly recirculate through the bloodstream to another lymph node. Therefore, regulation of a T-cell response is dependent upon the ability of cells to arrive in the correct location following chemokine gradients ("go" signal) as well as to receive appropriate T-cell receptor (TCR) activation signals upon cognate antigen recognition ("stop" signal). However, the mechanisms by which T cells regulate these go and stop signals remain unclear. We found that overexpression of the hematopoietic-specific RhoH protein in the presence of chemokine signals resulted in decreased Rap1-GTP and LFA-1 adhesiveness to ICAM-1, thus impairing T-cell chemotaxis; while in the presence of TCR signals, there were enhanced and sustained Rap1-GTP and LFA-1 activation as well as prolonged T:APC conjugates. RT-PCR analyses of activated CD4(+) T cells and live images of T-cell migration and immunological synapse (IS) formation revealed that functions of RhoH took place primarily at the levels of transcription and intracellular distribution. Thus, we conclude that RhoH expression provides a key molecular determinant that allows T cells to switch between sensing chemokine-mediated go signals and TCR-dependent stop signals.


Assuntos
Ativação Linfocitária , Linfócitos T/citologia , Fatores de Transcrição/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Humanos , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores de Quimiocinas/fisiologia
12.
J Immunol ; 185(11): 7057-66, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21037096

RESUMO

To exit blood vessels, most (∼80%) of the lumenally adhered monocytes and neutrophils crawl toward locations that support transmigration. Using intravital confocal microscopy of anesthetized mouse cremaster muscle, we separately examined the crawling and emigration patterns of monocytes and neutrophils in blood-perfused unstimulated or TNF-α-activated venules. Most of the interacting cells in microvessels are neutrophils; however, in unstimulated venules, a greater percentage of the total monocyte population is adherent compared with neutrophils (58.2 ± 6.1% versus 13.6 ± 0.9%, adhered/total interacting), and they crawl for significantly longer distances (147.3 ± 13.4 versus 61.8 ± 5.4 µm). Intriguingly, after TNF-α activation, monocytes crawled for significantly shorter distances (67.4 ± 9.6 µm), resembling neutrophil crawling. Using function-blocking Abs, we show that these different crawling patterns were due to CD11a/CD18 (LFA-1)- versus CD11b/CD18 (Mac-1)-mediated crawling. Blockade of either Mac-1 or LFA-1 revealed that both LFA-1 and Mac-1 contribute to monocyte crawling; however, the LFA-1-dependent crawling in unstimulated venules becomes Mac-1 dependent upon inflammation, likely due to increased expression of Mac-1. Mac-1 alone was responsible for neutrophil crawling in both unstimulated and TNF-α-activated venules. Consistent with the role of Mac-1 in crawling, Mac-1 block (compared with LFA-1) was also significantly more efficient in blocking TNF-α-induced extravasation of both monocytes and neutrophils in cremaster tissue and the peritoneal cavity. Thus, mechanisms underlying leukocyte crawling are important in regulating the inflammatory responses by regulating the numbers of leukocytes that transmigrate.


Assuntos
Movimento Celular/imunologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Antígeno de Macrófago 1/fisiologia , Monócitos/imunologia , Neutrófilos/imunologia , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos CD18/fisiologia , Citometria de Fluxo , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Contagem de Leucócitos , Antígeno-1 Associado à Função Linfocitária/biossíntese , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno de Macrófago 1/biossíntese , Antígeno de Macrófago 1/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia de Fluorescência , Monócitos/metabolismo , Monócitos/ultraestrutura , Ativação de Neutrófilo/imunologia , Neutrófilos/metabolismo , Neutrófilos/ultraestrutura , Fator de Necrose Tumoral alfa/administração & dosagem , Vênulas/imunologia , Vênulas/metabolismo , Vênulas/ultraestrutura
13.
J Immunol ; 183(10): 6460-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864611

RESUMO

Intracellular signals associated with or triggered by integrin ligation can control cell survival, differentiation, proliferation, and migration. Despite accumulating evidence that conformational changes regulate integrin affinity to its ligands, how integrin structure regulates signal transmission from the outside to the inside of the cell remains elusive. Using fluorescence resonance energy transfer, we addressed whether conformational changes in integrin Mac-1 are sufficient to transmit outside-in signals in human neutrophils. Mac-1 conformational activation induced by ligand occupancy or activating Ab binding, but not integrin clustering, triggered similar patterns of intracellular protein tyrosine phosphorylation, including Akt phosphorylation, and inhibited spontaneous neutrophil apoptosis, indicating that global conformational changes are critical for Mac-1-dependent outside-in signal transduction. In neutrophils and myeloid K562 cells, ligand ICAM-1 or activating Ab binding promoted switchblade-like extension of the Mac-1 extracellular domain and separation of the alpha(M) and beta(2) subunit cytoplasmic tails, two structural hallmarks of integrin activation. These data suggest the primacy of global conformational changes in the generation of Mac-1 outside-in signals.


Assuntos
Leucócitos Mononucleares/imunologia , Antígeno de Macrófago 1/imunologia , Neutrófilos/imunologia , Transdução de Sinais/imunologia , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Carcinógenos/farmacologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Linhagem Celular Tumoral , Humanos , Fatores Imunológicos/farmacologia , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-8/farmacologia , Leucemia/imunologia , Leucemia/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Antígeno de Macrófago 1/efeitos dos fármacos , Antígeno de Macrófago 1/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
14.
Blood Cells Mol Dis ; 42(3): 177-84, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19246218

RESUMO

Substituting the medium chloride with glucuronate or glutamate causes a rapid, 10 to 30-fold, increase in the binding of the monoclonal antibody, CBRM1/5, which recognizes the high-affinity conformation of the Mac-1 integrin. This change is reflected in functional adhesion assays that show increased adhesion to ICAM-1 coated beads. Blocking antibodies indicate that the increased adhesion is almost entirely due to Mac-1. The inhibitor NPPB (100 microM) reduces Cl(-) efflux into low Cl(-) medium by 75%, and blocks increased CBRM1/5 binding after stimulation with fMLP or TNF-alpha, but has no effect on the anion substitution induced increase in CBRM1/5 binding or adhesion to immobilized ICAM-1. Thus, changes in external anion composition, not internal chloride or increases in Cl(-) efflux, are responsible for Mac-1 activation. This effect is substantial. The percentage of Mac-1 in the high affinity state approaches 100% in glutamate and 50% in glucuronate, a far greater response than what is observed after stimulation with fMLP.


Assuntos
Cloretos/farmacologia , Antígeno de Macrófago 1/metabolismo , Neutrófilos/citologia , Anticorpos Monoclonais/metabolismo , Adesão Celular/efeitos dos fármacos , Cloretos/metabolismo , Líquido Extracelular/química , Glucuronatos/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Microesferas , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/metabolismo , Ligação Proteica/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA