Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Physiol ; 594(3): 687-97, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26661689

RESUMEN

KEY POINTS: The local arteriolar dilatation produced by contraction of skeletal muscle is dependent upon multiple signalling mechanisms. In addition to the many metabolic signals that mediate this vasodilatation, we show here that the extracellular matrix protein fibronectin also contributes to the response. This vasodilatory signal requires the heparin-binding matricryptic RWRPK sequence in the first type III repeat of fibrillar fibronectin. The fibronectin-dependent component of the integrated muscle contraction-dependent arteriolar vasodilatation is coupled through an endothelial cell-dependent signalling pathway. Recent studies in contracting skeletal muscle have shown that functional vasodilatation in resistance arterioles has an endothelial cell (EC)-dependent component, and, separately have shown that the extracellular matrix protein fibronectin (FN) contributes to functional dilatation in these arterioles. Here we test the hypotheses that (i) the matricryptic heparin-binding region of the first type III repeat of fibrillar FN (FNIII1H) mediates vasodilatation, and (ii) this response is EC dependent. Engineered FN fragments with differing (defined) heparin- and integrin-binding capacities were applied directly to resistance arterioles in cremaster muscles of anaesthetized (pentobarbital sodium, 65 mg kg(-1)) mice. Both FNIII1H,8-10 and FNIII1H induced dilatations (12.2 ± 1.7 µm, n = 12 and 17.2 ± 2.4 µm, n = 14, respectively) whereas mutation of the active sequence (R(613) WRPK) of the heparin binding region significantly diminished the dilatation (3.2 ± 1.8 µm, n = 10). Contraction of skeletal muscle fibres via electrical field stimulation produced a vasodilatation (19.4 ± 1.2 µm, n = 12) that was significantly decreased (to 7.0 ± 2.7 µm, n = 7, P < 0.05) in the presence of FNIII1Peptide 6, which blocks extracellular matrix (ECM) FN and FNIII1H signalling. Furthermore, FNIII1H,8-10 and FNIII1H applied to EC-denuded arterioles failed to produce any dilatation indicating that endothelium was required for the response. Finally, FNIII1H significantly increased EC Ca(2+) (relative fluorescence 0.98 ± 0.02 in controls versus 1.12 ± 0.05, n = 17, P < 0.05). Thus, we conclude that ECM FN-dependent vasodilatation is mediated by the heparin-binding (RWRPK) sequence of FNIII1 in an EC-dependent manner. Importantly, blocking this signalling sequence decreased the dilatation to skeletal muscle contraction, indicating that there is a physiological role for this FN-dependent mechanism.


Asunto(s)
Arteriolas/fisiología , Matriz Extracelular/fisiología , Fibronectinas/fisiología , Músculo Esquelético/fisiología , Animales , Calcio/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Heparina/metabolismo , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Péptidos/fisiología , Unión Proteica , Proteínas Recombinantes de Fusión , Vasodilatación/fisiología
2.
Am J Physiol Heart Circ Physiol ; 311(4): H1063-H1071, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521419

RESUMEN

Endothelial cells (EC) respond to mechanical forces such as shear stress in a variety of ways, one of which is cytoskeletal realignment in the direction of flow. Our earlier studies implicated the extracellular matrix protein fibronectin in mechanosensory signaling to ECs in intact arterioles, via a signaling pathway dependent on the heparin-binding region of the first type III repeat of fibrillar fibronectin (FNIII1H). Here we test the hypothesis that FNIII1H is required for EC stress fiber realignment under flow. Human umbilical vein ECs (HUVECs) exposed to defined flow conditions were used as a well-characterized model of this stress fiber alignment response. Our results directly implicate FNIII1H in realignment of stress fibers in HUVECs and, importantly, show that the matricryptic heparin-binding RWRPK sequence located in FNIII1 is required for the response. Furthermore, we show that flow-mediated stress fiber realignment in ECs adhered via α5ß1-integrin-specific ligands does not occur in the absence of FHIII1H, whereas, in contrast, αvß3-integrin-mediated stress fiber realignment under flow does not require FNIII1H. Our findings thus indicate that there are two separate mechanosignaling pathways mediating the alignment of stress fibers after exposure of ECs to flow, one dependent on αvß3-integrins and one dependent on FNIII1H. This study strongly supports the conclusion that the RWRPK region of FNIII1H may have broad capability as a mechanosensory signaling site.


Asunto(s)
Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Mecanotransducción Celular , Estrés Mecánico , Células Endoteliales/fisiología , Heparina , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Microscopía Fluorescente
3.
J Physiol ; 593(21): 4699-711, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26314391

RESUMEN

The focus of this review is on local mechanisms modifying arteriolar resistance to match blood flow to metabolism. In skeletal muscle many local mediators are known, including K(+) , nitric oxide (NO), purines and prostaglandins. Each accounts for about 30% of the response; it is widely held that these act redundantly: this concept awaits systematic testing. Understanding signal integration also requires consideration of microvascular network morphology in relation to local communication pathways between endothelial and smooth muscle cells (which are critical for many local responses, including dilatation to skeletal muscle contraction) and in relation to the spread of vasodilator signals up- and downstream throughout the network. Mechanisms mediating the spread of dilatation from local to remote sites have been well studied using acetylcholine (ACh), but remote dilatations to contraction of skeletal muscle fibres also occur. Importantly, these mechanisms clearly differ from those initiated by ACh, but much remains undefined. Furthermore, capillaries contribute to metabolic dilatation as they dilate arterioles directly upstream in response to vasoactive agents or contraction of adjacent muscle fibres. Given the dispersed arrangement of motor units, precise matching of flow to metabolism is not attainable unless signals are initiated only by 'active' capillaries. As motor units are recruited, signals that direct blood flow towards these active fibres will eventually be supported by local and spreading responses in the arterioles associated with those fibres. Thus, mechanisms of integration of vasodilator signalling across elements of the microvasculature remain an important area of focus for new studies.


Asunto(s)
Hiperemia/metabolismo , Microvasos/metabolismo , Acetilcolina/metabolismo , Animales , Humanos , Hiperemia/fisiopatología , Canales KATP/metabolismo , Microvasos/fisiología , Óxido Nítrico/metabolismo
4.
J Biol Chem ; 289(47): 33054-65, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25320085

RESUMEN

Disruption of endothelial cell-cell contact is a key event in many cardiovascular diseases and a characteristic of pathologically activated vascular endothelium. The CCM (cerebral cavernous malformation) family of proteins (KRIT1 (Krev-interaction trapped 1), PDCD10, and CCM2) are critical regulators of endothelial cell-cell contact and vascular homeostasis. Here we show novel regulation of vascular endothelial growth factor (VEGF) signaling in KRIT1-depleted endothelial cells. Loss of KRIT1 and PDCD10, but not CCM2, increases nuclear ß-catenin signaling and up-regulates VEGF-A protein expression. In KRIT1-depleted cells, increased VEGF-A levels led to increased VEGF receptor 2 (VEGFR2) activation and subsequent alteration of cytoskeletal organization, migration, and barrier function and to in vivo endothelial permeability in KRIT1-deficient animals. VEGFR2 activation also increases ß-catenin phosphorylation but is only partially responsible for KRIT1 depletion-dependent disruption of cell-cell contacts. Thus, VEGF signaling contributes to modifying endothelial function in KRIT1-deficient cells and microvessel permeability in Krit1(+/-) mice; however, VEGF signaling is likely not the only contributor to disrupted endothelial cell-cell contacts in the absence of KRIT1.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Permeabilidad Capilar , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Bovinos , Comunicación Celular , Permeabilidad de la Membrana Celular , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteína KRIT1 , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Fosforilación , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/metabolismo
5.
J Physiol ; 592(12): 2575-90, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24687580

RESUMEN

Adenosine (ADO) is an endogenous vasodilatory purine widely recognized to be a significant contributor to functional hyperaemia. Despite this, many aspects of the mechanisms by which ADO induces dilation in small resistance arterioles are not established, or appear contradictory. These include: identification of the primary receptor subtype; its location on endothelial (EC) or vascular smooth muscle cells; whether ADO acts on KATP channels in these resistance vessels; and the contribution of cAMP/protein kinase A (PKA) signalling to the response. In intravital microscopy studies of intact or EC-denuded skeletal muscle arterioles, we show that ADO acts via A2A receptors located on ECs to produce vasodilation via activation of KATP channels located on vascular smooth muscle cells. Importantly, we found that the signalling pathway involves cAMP as expected, but that a requirement for PKA activation is demonstrable only if the vessel is not pre-exposed to ADO. That is, PKA-dependent signalling varies with pre-exposure to ADO. Further, we show that PKA activation alone is not sufficient to dilate these arterioles; an additional EC calcium-dependent signalling mechanism is required for vasodilation to ADO. The ability of arterioles in situ to respond to occupancy of a specific receptor by utilizing different cell signalling pathways under different conditions to produce the same response allows the arteriole to respond to key homeostatic requirements using more than a single signalling mechanism. Clearly, this is likely to be physiologically advantageous, but the role for this signalling flexibility in the integrated arteriolar response that underlies functional hyperaemia will require further exploration.


Asunto(s)
Adenosina/fisiología , Arteriolas/fisiología , Endotelio Vascular/fisiología , Receptor de Adenosina A2A/fisiología , Vasodilatación/fisiología , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células Endoteliales/fisiología , Hiperemia/fisiopatología , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/fisiología , Resistencia Vascular
6.
Microcirculation ; 20(8): 679-92, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23701383

RESUMEN

Arterioles, capillaries, and venules all actively change their cellular functions and phenotypes during inflammation in ways that are essential for maintenance of homeostasis and self-defense, and are also associated with many inflammatory disorders. ECs, together with pericytes and ECM proteins, can regulate blood flow, the coagulation cascade, fluid and solute exchange, and leukocyte trafficking. While capillary and venular functions in inflammation are well characterized, the arteriolar contribution to inflammation has only recently come into focus. Arterioles differ from venules in structure, EC morphology, shear environment, expression, and distribution of surface ligands; hence, regulation and function of arteriolar wall cells during inflammation may also be distinct from venules. Recent work indicates that in response to proinflammatory stimuli, arterioles alter barrier function, and support leukocyte and platelet interactions through upregulation of adhesion molecules. This suggests that in addition to their role in blood flow regulation, arterioles may also participate in inflammatory responses. In this review, we will discuss mechanisms that characterize arteriolar responses to proinflammatory stimuli. We will detail how distinct arteriolar features contribute to regulation of barrier function and leukocyte-EC interactions in inflammation, and further highlight the potential priming effects of arteriolar responses on venular function and progression of inflammatory responses.


Asunto(s)
Arteriolas/metabolismo , Plaquetas/metabolismo , Comunicación Celular , Inflamación/metabolismo , Leucocitos/metabolismo , Animales , Arteriolas/patología , Arteriolas/fisiopatología , Plaquetas/patología , Moléculas de Adhesión Celular/biosíntesis , Humanos , Inflamación/patología , Inflamación/fisiopatología , Leucocitos/patología , Regulación hacia Arriba
7.
Arterioscler Thromb Vasc Biol ; 32(11): 2702-10, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22922958

RESUMEN

OBJECTIVE: The regulation of vascular permeability, leukocyte trafficking, and the integrity of endothelial cell-cell contacts are closely linked by a complex mechanism of interregulation. Here, we investigate the role of Krev interaction-trapped 1 (KRIT1), an adherens junction accessory protein required for cell-cell junction stability, in regulating these vascular functions. METHODS AND RESULTS: Krit1(+/-) mice exhibited an enhanced edematous response to the complex inflammatory stimuli found in the passive K/BxN model of inflammatory arthritis and the murine air pouch model, yet leukocyte infiltration was unchanged. Correspondingly, reduced KRIT1 expression increased baseline arteriole and venule permeability 2-fold over that of wild-type littermates, as measured by intravital microscopy of the intact cremaster muscle vascular network, but this increase was not accompanied by increased leukocyte extravasation or activation. Direct stimulation with tumor necrosis factor-α induced increased permeability in wild-type mice, but surprisingly no increase over baseline levels was observed in Krit1(+/-) mice, despite extensive leukocyte activation. Finally, adoptive transfer of Krit1(+/-) bone marrow failed to increase permeability in wild-type mice. However, reduced expression of KRIT1 in the hematopoietic lineage dampened the differences observed in baseline permeability. CONCLUSIONS: Taken together, our data indicate an integral role for KRIT1 in microvessel homeostasis and the vascular response to inflammation.


Asunto(s)
Arteriolas/metabolismo , Artritis/metabolismo , Permeabilidad Capilar , Edema/metabolismo , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Vénulas/metabolismo , Animales , Arteriolas/inmunología , Artritis/genética , Artritis/inmunología , Artritis/patología , Trasplante de Médula Ósea , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Edema/genética , Edema/inmunología , Edema/patología , Homeostasis , Humanos , Mediadores de Inflamación/metabolismo , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/patología , Proteína KRIT1 , Rodamiento de Leucocito , Leucocitos/inmunología , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Tiempo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Vénulas/inmunología
8.
J Immunol ; 184(9): 5242-52, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20363969

RESUMEN

Leukocyte transmigration occurs at specific locations (portals) on the endothelium, but the nature of these portals is not clear. Using intravital confocal microscopy of anesthetized mouse cremaster muscle in combination with immunofluorescence labeling, we showed that in microvessels transmigration is mainly junctional and preferentially occurs at tricellular endothelial junctional regions. Our data suggest that enrichment of ICAM-1 near approximately 43% of these junctions makes these locations preferred for transmigration by signaling the location of a nearby portal, as well as preparing the endothelial cell (EC) junctions, to accommodate leukocyte passage. Blockade of the extracellular domain of the ICAM-1 significantly reduced transmigration (by 68.8 + or - 4.5%) by reducing the ability of leukocytes to get to these portals. In contrast, blockade of the cytoplasmic tail of ICAM-1 reduced transmigration (by 71.1 + or - 7.0%) by disabling VE-cadherin rearrangement. Importantly, venular convergences are optimally equipped to support leukocyte transmigration. Differences in EC morphology result in a significantly higher number of tricellular junctions in convergences compared with straight venular regions (20.7 + or - 1.2 versus 12.43 + or - 1.1/6000 microm(2), respectively). Consequently, leukocyte adhesion and transmigration are significantly higher in convergences compared with straight regions (1.6- and 2.6-fold, respectively). Taken together, these data identify an important role for EC morphology and expression patterns of ICAM-1 in leukocyte transmigration.


Asunto(s)
Uniones Adherentes/inmunología , Quimiotaxis de Leucocito/inmunología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Rodamiento de Leucocito/inmunología , Transducción de Señal/inmunología , Uniones Adherentes/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/inmunología , Endotelio Vascular/inmunología , Molécula 1 de Adhesión Intercelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Microvasos/inmunología , Microvasos/metabolismo , Datos de Secuencia Molecular , Vénulas/citología , Vénulas/inmunología , Vénulas/metabolismo
9.
J Immunol ; 185(11): 7057-66, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21037096

RESUMEN

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.


Asunto(s)
Movimiento Celular/inmunología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Antígeno de Macrófago-1/fisiología , Monocitos/inmunología , Neutrófilos/inmunología , Animales , Anticuerpos Bloqueadores/farmacología , Antígenos CD18/fisiología , Citometría de Flujo , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Recuento de Leucocitos , Antígeno-1 Asociado a Función de Linfocito/biosíntesis , Antígeno-1 Asociado a Función de Linfocito/inmunología , Antígeno de Macrófago-1/biosíntesis , Antígeno de Macrófago-1/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente , Monocitos/metabolismo , Monocitos/ultraestructura , Activación Neutrófila/inmunología , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Factor de Necrosis Tumoral alfa/administración & dosificación , Vénulas/inmunología , Vénulas/metabolismo , Vénulas/ultraestructura
10.
Am J Physiol Cell Physiol ; 301(4): C804-13, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21653902

RESUMEN

Activated neutrophils interacting with the vessel wall can alter vascular permeability to macromolecules such as albumin via release of various secretion products that induce changes in the endothelial monolayer. In the current work we used cremaster microvessels of anesthetized mice to show that, in addition to this paracrine mechanism, leukocyte ligation of endothelial ICAM-1 directly activates endothelial cell (EC) signaling, altering EC permeability to albumin [i.e., solute permeability (P(s))]. We show that antibody cross-linking of surface ICAM-1 in intact microvessels is sufficient to increase P(s) even in the absence of interacting leukocytes. Unstimulated arterioles do not support leukocyte-EC interactions, but despite this, antibody ligation of ICAM-1 in these vessels induced a twofold increase in P(s). Similarly, in venules that were depleted of interacting neutrophils, P(s) was decreased to below resting levels and was restored by ligation of ICAM-1. Use of function-blocking antibodies to separately block leukocyte rolling or adhesion under unstimulated or TNF-α-activated conditions established that both rolling and adhered leukocytes contribute to P(s) regulation in situ. Both rolling and adhesion activated EC-dependent signaling mechanisms that increased P(s). ICAM-1 ligation with primary antibody alone or primary followed by secondary antibodies showed that regulation of P(s) is directly dependent on the degree of ICAM-1 clustering. Under physiological versus inflamed conditions, respectively, this ICAM-1 clustering-dependent regulation of P(s) switches from PKC dependent and Src independent to Src dependent and PKC independent. This study thus identifies a new mechanism by which antiadhesion treatment may constitute a potential therapy for tissue edema.


Asunto(s)
Albúminas/fisiología , Permeabilidad Capilar/fisiología , Molécula 1 de Adhesión Intercelular/metabolismo , Rodamiento de Leucocito/fisiología , Leucocitos/fisiología , Animales , Adhesión Celular , Regulación de la Expresión Génica , Leucocitos/citología , Ratones , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
11.
Circ Res ; 102(3): 372-9, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18032733

RESUMEN

During exercise, local mechanisms in tissues cause arterioles to rapidly dilate to increase blood flow to tissues to meet the metabolic demands of contracting muscle. Despite decades of study, the mechanisms underlying this important aspect of blood flow control are still far from clear. We now report a novel mechanism wherein fibronectin fibrils in connective tissue matrices transduce signals from contracting skeletal muscle to local blood vessels to increase blood flow. Using intravital microscopy, we show that local vasodilation in response to skeletal muscle contraction is specifically inhibited by an antibody that recognizes the matricryptic site in the first type III repeat of fibronectin (FNIII-1). In the absence of skeletal muscle contraction, direct application of FNIII-1-containing fibronectin fragments to cremaster muscle arterioles in situ, triggered a rapid, specific, and reversible local dilation that was mediated by nitric oxide and required the cryptic, heparin-binding sequence of FNIII-1. Furthermore, application of function-blocking FNIII-1 peptides to cremaster muscle arterioles rapidly and specifically decreased their diameter, indicating that the matricryptic site of fibronectin also contributes to resting vascular tone. Alexa fluor 488-labeled fibronectin, administered intravenously, was rapidly assembled into elongated, branching fibrils in the extracellular matrix of intact cremaster muscle, demonstrating active polymerization of fibronectin in areas adjacent to blood vessels. Together, these data provide the first evidence that a matricryptic, heparin-binding site within fibronectin fibrils of adult connective tissue plays a dynamic role in regulating both vascular responses and vascular tone.


Asunto(s)
Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/irrigación sanguínea , Péptidos/metabolismo , Transducción de Señal/fisiología , Vasodilatación/fisiología , Animales , Arteriolas/fisiología , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Velocidad del Flujo Sanguíneo/fisiología , Cricetinae , Cricetulus , Fibronectinas/farmacología , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/fisiología , Óxido Nítrico/metabolismo , Péptidos/farmacología , Condicionamiento Físico Animal/fisiología , Vasodilatación/efectos de los fármacos
12.
Microcirculation ; 16(6): 508-20, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19468960

RESUMEN

OBJECTIVE: Variation in expression of adhesion molecules plays a key role in regulating leukocyte behavior, but the contribution of fluid shear to these interactions cannot be ignored. Here, we dissected the effects of each of these factors on leukocyte behavior in different venular regions. MATERIALS AND METHODS: Leukocyte behavior was quantified in blood-perfused microvascular networks in anesthetized mouse cremaster muscle, using intravital confocal microscopy. ICAM-1 expression and fluid shear rate were quantified by using ICAM-1 fluorescent labeling, fluorescent particle tracking, and computational fluid dynamics. RESULTS: Tumor necrosis factor alpha induced an increase in ICAM-1 expression and abolished the differences observed among control venules of different sizes. Consequently, leukocyte adhesion was increased to a similar level across all vessel sizes [5.1+/-0.46 leukocytes/100 microm vs. 2.1+/-0.47 (control)], but remained significantly higher in venular convergences (7.8+/-0.4). Leukocyte transmigration occurred primarily in the smallest venules and venular convergences (23.9+/-5.1 and 31.9+/-2.7 leukocytes/10,000 microm(2) tissue, respectively). In venular convergences, the two inlet vessels are predicted to create a region of low velocity, increasing leukocyte adhesion probability. CONCLUSIONS: In straight regions of different-sized venules, the variability in ICAM-1 expression accounts for the differences in leukocyte behavior; in converging regions, fluid shear potentially has a greater effect on leukocyte endothelial cell interactions.


Asunto(s)
Molécula 1 de Adhesión Intercelular/fisiología , Leucocitos/fisiología , Vénulas/anatomía & histología , Vénulas/fisiología , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Hemorreología , Leucocitos/citología , Leucocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Músculo Esquelético/irrigación sanguínea , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/farmacología
13.
Sci Rep ; 7(1): 8296, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811547

RESUMEN

The intracellular scaffold KRIT1/CCM1 is an established regulator of vascular barrier function. Loss of KRIT1 leads to decreased microvessel barrier function and to the development of the vascular disorder Cerebral Cavernous Malformation (CCM). However, how loss of KRIT1 causes the subsequent deficit in barrier function remains undefined. Previous studies have shown that loss of KRIT1 increases the production of reactive oxygen species (ROS) and exacerbates vascular permeability triggered by several inflammatory stimuli, but not TNF-α. We now show that endothelial ROS production directly contributes to the loss of barrier function in KRIT1 deficient animals and cells, as targeted antioxidant enzymes reversed the increase in permeability in KRIT1 heterozygous mice as shown by intravital microscopy. Rescue of the redox state restored responsiveness to TNF-α in KRIT1 deficient arterioles, but not venules. In vitro, KRIT1 depletion increased endothelial ROS production via NADPH oxidase signaling, up-regulated Nox4 expression, and promoted NF-κB dependent promoter activity. Recombinant yeast avenanthramide I, an antioxidant and inhibitor of NF-κB signaling, rescued barrier function in KRIT1 deficient cells. However, KRIT1 depletion blunted ROS production in response to TNF-α. Together, our data indicate that ROS signaling is critical for the loss of barrier function following genetic deletion of KRIT1.


Asunto(s)
Endotelio/metabolismo , Proteína KRIT1/deficiencia , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Transducción de Señal , Animales , Antioxidantes/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Regulación de la Expresión Génica , Proteína KRIT1/genética , Proteína KRIT1/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Biorheology ; 42(5): 363-83, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16308467

RESUMEN

The aim of this study was to characterize the distribution of adherent leukocytes in branched venular convergences in vivo. Intravital microscopy was used to obtain video images of leukocyte adhesion in multiple branched sites in mouse cremaster muscle, during the mild inflammatory response induced by surgical preparation. The average number of cells/vessel length was obtained over several minutes for seven venular convergences with varying geometrical configurations. Results from this study demonstrate a strong tendency of leukocytes to adhere at junctional points between converging vessels. Different vessel configurations were studied and results were shown to be insensitive to precise vessel geometry. Thus, in post-capillary venules, leukocytes are most likely to adhere at points between converging vessels, regardless of the precise geometrical properties or configuration of the vessels. Hydrodynamic mechanisms due to flow behavior through convergences likely play a significant role in determining locations of cellular adhesion. Future work should concentrate on quantifying the relative contributions of hydrodynamic and biochemical mechanisms to aid in understanding disease processes and development of treatments or therapeutics.


Asunto(s)
Rodamiento de Leucocito , Neutrófilos , Vénulas/inmunología , Animales , Adhesión Celular , Endotelio Vascular/inmunología , Inflamación , Recuento de Leucocitos , Ratones , Microscopía por Video , Músculo Esquelético/irrigación sanguínea , Flujo Sanguíneo Regional
15.
Tissue Barriers ; 3(1-2): e985954, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25838987

RESUMEN

Vascular permeability is a vital function of the circulatory system that is regulated in large part by the limited flux of solutes, water, and cells through the endothelial cell layer. One major pathway through this barrier is via the inter-endothelial junction, which is driven by the regulation of cadherin-based adhesions. The endothelium also forms attachments with surrounding proteins and cells via 2 classes of adhesion molecules, the integrins and IgCAMs. Integrins and IgCAMs propagate activation of multiple downstream signals that potentially impact cadherin adhesion. Here we discuss the known contributions of integrin and IgCAM signaling to the regulation of cadherin adhesion stability, endothelial barrier function, and vascular permeability. Emphasis is placed on known and prospective crosstalk signaling mechanisms between integrins, the IgCAMs- ICAM-1 and PECAM-1, and inter-endothelial cadherin adhesions, as potential strategic signaling nodes for multipartite regulation of cadherin adhesion.

16.
PLoS One ; 10(6): e0128378, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26091091

RESUMEN

P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) play important roles in mediating the inflammatory cascade. Selectin kinetics, together with neutrophil hydrodynamics, regulate the fundamental adhesion cascade of cell tethering and rolling on the endothelium. The current study uses the Multiscale Adhesive Dynamics computational model to simulate, for the first time, the tethering and rolling behavior of pseudopod-containing neutrophils as mediated by P-selectin/PSGL-1 bonds. This paper looks at the effect of including P-selectin/PSGL-1 adhesion kinetics. The parameters examined included the shear rate, adhesion on-rate, initial neutrophil position, and receptor number sensitivity. The outcomes analyzed included types of adhesive behavior observed, tether rolling distance and time, number of bonds formed during an adhesive event, contact area, and contact time. In contrast to the hydrodynamic model, P-selectin/PSGL-1 binding slows the neutrophil's translation in the direction of flow and causes the neutrophil to swing around perpendicular to flow. Several behaviors were observed during the simulations, including tethering without firm adhesion, tethering with downstream firm adhesion, and firm adhesion upon first contact with the endothelium. These behaviors were qualitatively consistent with in vivo data of murine neutrophils with pseudopods. In the simulations, increasing shear rate, receptor count, and bond formation rate increased the incidence of firm adhesion upon first contact with the endothelium. Tethering was conserved across a range of physiological shear rates and was resistant to fluctuations in the number of surface PSGL-1 molecules. In simulations where bonding occurred, interaction with the side of the pseudopod, rather than the tip, afforded more surface area and greater contact time with the endothelial wall.


Asunto(s)
Simulación por Computador , Modelos Biológicos , Neutrófilos/fisiología , Algoritmos , Adhesión Celular , Endotelio Vascular , Rodamiento de Leucocito , Selectinas/metabolismo , Estrés Mecánico
17.
J Exp Med ; 209(7): 1349-62, 2012 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-22711877

RESUMEN

The efficient trafficking of immune cells into peripheral nonlymphoid tissues is key to enact their protective functions. Despite considerable advances in our understanding of cell migration in secondary lymphoid organs, real-time leukocyte recruitment into inflamed tissues is not well characterized. The conventional multistep paradigm of leukocyte extravasation depends on CD18 integrin-mediated events such as rapid arrest and crawling on the surface of the endothelium and transmigration through the endothelial layer. Using enhanced three-dimensional detection of fluorescent CD18 fusion proteins in a newly developed knockin mouse, we report that extravasating leukocytes (neutrophils, monocytes, and T cells) show delayed uropod detachment and become extremely elongated before complete transmigration across the endothelium. Additionally, these cells deposit CD18(+) microparticles at the subendothelial layer before retracting the stretched uropod. Experiments with knockout mice and blocking antibodies reveal that the uropod elongation and microparticle formation are the result of LFA-1-mediated adhesion and VLA-3-mediated cell migration through the vascular basement membrane. These findings suggest that uropod elongation is a final step in the leukocyte extravasation cascade, which may be important for precise regulation of leukocyte recruitment into inflamed tissues.


Asunto(s)
Extensiones de la Superficie Celular/fisiología , Leucocitos/fisiología , Migración Transendotelial y Transepitelial/fisiología , Vasculitis/metabolismo , Animales , Antígenos CD18/genética , Antígenos CD18/metabolismo , Adhesión Celular/genética , Adhesión Celular/fisiología , Extensiones de la Superficie Celular/genética , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina alfa3beta1/deficiencia , Integrina alfa3beta1/genética , Leucocitos/metabolismo , Leucocitos/ultraestructura , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Microscopía Fluorescente/métodos , Neutrófilos/metabolismo , Neutrófilos/fisiología , Neutrófilos/ultraestructura , Linfocitos T/metabolismo , Linfocitos T/fisiología , Linfocitos T/ultraestructura , Migración Transendotelial y Transepitelial/genética , Vasculitis/genética
18.
Ann Biomed Eng ; 37(5): 913-26, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19280342

RESUMEN

The blood vessel diameter is often measured in microcirculation studies to quantify the effects of various stimuli. Intravital video microscopy is used to measure the change in vessel diameter by first recording the video and analyzing it using electronic calipers or by using image shearing technique. Manual measurement using electronic calipers or image shearing is time-consuming and prone to measurement error, and automated measurement can serve as an alternative that is faster and more reliable. In this paper, a new feature-based tracking algorithm is presented for automatically measuring diameter of vessels in intravital video microscopy image sequences. Our method tracks the vessel diameter throughout the entire image sequence once the diameter is marked in the first image. The parameters were calibrated using the intravital videos with manual ground truth measurements. The experiment with 10 synthetic videos and 20 intravital microscopy videos, including 10 fluorescence confocal and 10 non-confocal transmission, shows that the measurement can be performed accurately.


Asunto(s)
Algoritmos , Vasos Sanguíneos/anatomía & histología , Microscopía por Video/métodos , Vasos Sanguíneos/ultraestructura , Procesamiento de Imagen Asistido por Computador , Microcirculación , Microscopía Electrónica de Transmisión , Microscopía Fluorescente/métodos , Modelos Cardiovasculares , Tamaño de los Órganos
19.
Am J Physiol Heart Circ Physiol ; 295(3): H969-H977, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18641276

RESUMEN

Two key characteristics of the inflammatory response are the recruitment of leukocytes to inflamed tissue as well as changes in vessel permeability. We explored the relationship between these two processes using intravital confocal microscopy in cremasters of anesthetized (65 mg/kg Nembutal ip) mice. We provide direct evidence that intercellular adhesion molecule-1 (ICAM-1) links leukocyte-endothelial cell interactions and changes in solute permeability (Ps). Importantly, we show that arterioles, not just venules, respond to proinflammatory stimuli, thus contributing to microvascular exchange. We identified two independent, ICAM-1-mediated pathways regulating Ps. Under control conditions in wild-type (WT) mice, there is a constitutive PKC-dependent pathway (Ps = 1.0 +/- 0.10 and 2.2 +/- 0.46 x 10(-6) cm/s in arterioles and venules, respectively), which was significantly reduced in ICAM-1 knockout (KO) mice (Ps = 0.54 +/- 0.07 and 0.77 +/- 0.11 x 10(-6) cm/s). The PKC inhibitor bisindolylmaleimid l (1 micromol/l in 0.01% DMSO) decreased P(s) in WT mice to levels similar to those in ICAM-1 KO mice. Likewise, a PKC activator (phorbol-12-myristate-acetate; 1 micromol/l in 0.01% DMSO) successfully restored Ps in ICAM-1 KO vessels to be not different from that of the WT controls. On the other hand, during TNF-alpha-induced inflammation, Ps in WT mice was significantly increased (2-fold in venules and 2.5-fold in arterioles) in a Src-dependent and PKC-independent manner. The blockade of Src (PP2; 2 micromol/l in 0.01% DMSO) but not PKC significantly reduced the TNF-alpha-dependent increase in Ps. We conclude that ICAM-1 plays an essential role in the regulation of Ps in microvessels and that there are two separate (constitutive and inducible) signaling pathways that regulate permeability under normal and inflamed conditions.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/fisiología , Molécula 1 de Adhesión Intercelular/fisiología , Leucocitos/fisiología , Transducción de Señal/fisiología , Animales , Antígenos CD18/fisiología , Capilares/patología , Inhibidores Enzimáticos/farmacología , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/fisiología
20.
Ann Biomed Eng ; 36(4): 580-5, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18317930

RESUMEN

A recent analytical solution of the three-dimensional Stokes flow through a bumpy tube predicts that for a given bump area, there exists an optimal circumferential wavenumber which minimizes flow resistance. This study uses measurements of microvessel endothelial cell morphology to test whether this prediction holds in the microvasculature. Endothelial cell (EC) morphology was measured in blood perfused in situ microvessels in anesthetized mice using confocal intravital microscopy. EC borders were identified by immunofluorescently labeling the EC surface molecule ICAM-1 which is expressed on the surface but not in the EC border regions. Comparison of this theory with extensive in situ measurements of microvascular EC geometry in mouse cremaster muscle using intravital microscopy reveals that the spacing of EC nuclei in venules ranging from 27 to 106 microm in diameter indeed lies quite close to this predicted optimal configuration. Interestingly, arteriolar ECs are configured to minimize flow resistance not in the resting state, but at the dilated vessel diameter. These results raise the question of whether less organized circulatory systems, such as that found in newly formed solid tumors or in the developing embryo, may deviate from the optimal bump spacing predicted to minimize flow resistance.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Mecanotransducción Celular/fisiología , Microcirculación/citología , Microcirculación/fisiología , Modelos Cardiovasculares , Animales , Tamaño de la Célula , Células Cultivadas , Simulación por Computador , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Resistencia Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA