Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
FEBS J ; 290(4): 1078-1095, 2023 02.
Article in English | MEDLINE | ID: mdl-36107440

ABSTRACT

Loss of Krev interaction-trapped-1 (KRIT1) expression leads to the development of cerebral cavernous malformations (CCM), a disease in which abnormal blood vessel formation compromises the structure and function of the blood-brain barrier. The role of KRIT1 in regulating endothelial function is well-established. However, several studies have suggested that KRIT1 could also play a role in regulating nonendothelial cell types and, in particular, immune cells. In this study, we generated a mouse model with neutrophil-specific deletion of KRIT1 in order to investigate the effect of KRIT1 deficiency on neutrophil function. Neutrophils isolated from adult Ly6Gtm2621(cre)Arte Krit1flox/flox mice had a reduced ability to attach and spread on the extracellular matrix protein fibronectin and exhibited a subsequent increase in migration. However, adhesion to and migration on ICAM-1 was unchanged. In addition, we used a monomeric, fluorescently-labelled fragment of fibronectin to show that integrin activation is reduced in the absence of KRIT1 expression, though ß1 integrin expression appears unchanged. Finally, neutrophil migration in response to lipopolysaccharide-induced inflammation in the lung was decreased, as shown by reduced cell number and myeloperoxidase activity in lavage samples from Krit1PMNKO mice. Altogether, we show that KRIT1 regulates neutrophil adhesion and migration, likely through regulation of integrin activation, which can lead to altered inflammatory responses in vivo.


Subject(s)
Cell Adhesion , Cell Movement , KRIT1 Protein , Neutrophils , Animals , Mice , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/genetics , Cell Movement/physiology , Fibronectins , Integrin beta1/metabolism , KRIT1 Protein/metabolism , Microtubule-Associated Proteins/metabolism , Neutrophils/metabolism , Proto-Oncogene Proteins/metabolism
2.
Antioxid Redox Signal ; 38(7-9): 496-528, 2023 03.
Article in English | MEDLINE | ID: mdl-36047808

ABSTRACT

Significance: KRIT1 (Krev interaction trapped 1) is a scaffolding protein that plays a critical role in vascular morphogenesis and homeostasis. Its loss-of-function has been unequivocally associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease of genetic origin characterized by defective endothelial cell-cell adhesion and ensuing structural alterations and hyperpermeability in brain capillaries. KRIT1 contributes to the maintenance of endothelial barrier function by stabilizing the integrity of adherens junctions and inhibiting the formation of actin stress fibers. Recent Advances: Among the multiple regulatory mechanisms proposed so far, significant evidence accumulated over the past decade has clearly shown that the role of KRIT1 in the stability of endothelial barriers, including the blood-brain barrier, is largely based on its involvement in the complex machinery governing cellular redox homeostasis and responses to oxidative stress and inflammation. KRIT1 loss-of-function has, indeed, been demonstrated to cause an impairment of major redox-sensitive mechanisms involved in spatiotemporal regulation of cell adhesion and signaling, which ultimately leads to decreased cell-cell junction stability and enhanced sensitivity to oxidative stress and inflammation. Critical Issues: This review explores the redox mechanisms that influence endothelial cell adhesion and barrier function, focusing on the role of KRIT1 in such mechanisms. We propose that this supports a novel model wherein redox signaling forms the common link between the various pathogenetic mechanisms and therapeutic approaches hitherto associated with CCM disease. Future Directions: A comprehensive characterization of the role of KRIT1 in redox control of endothelial barrier physiology and defense against oxy-inflammatory insults will provide valuable insights into the development of precision medicine strategies. Antioxid. Redox Signal. 38, 496-528.


Subject(s)
Hemangioma, Cavernous, Central Nervous System , Humans , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/metabolism , Endothelial Cells/metabolism , Signal Transduction , Oxidation-Reduction , Inflammation , Microtubule-Associated Proteins/metabolism , KRIT1 Protein/metabolism
3.
Front Cardiovasc Med ; 9: 954780, 2022.
Article in English | MEDLINE | ID: mdl-35898265

ABSTRACT

Recent advances have steadily increased the number of proteins and pathways known to be involved in the development of cerebral cavernous malformation (CCM). Our ability to synthesize this information into a cohesive and accurate signaling model is limited, however, by significant gaps in our knowledge of how the core CCM proteins, whose loss of function drives development of CCM, are regulated. Here, we review what is known about the regulation of the three core CCM proteins, the scaffolds KRIT1, CCM2, and CCM3, with an emphasis on binding interactions and subcellular location, which frequently control scaffolding protein function. We highlight recent work that challenges the current model of CCM complex signaling and provide recommendations for future studies needed to address the large number of outstanding questions.

4.
J Cell Sci ; 135(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-34918736

ABSTRACT

Krev-interaction trapped protein 1 (KRIT1) is an endothelial scaffold protein that promotes adherens junction (AJ) stability. The precise mechanism by which KRIT1 promotes barrier stabilization is unclear. We tested the ability of a panel of KRIT1 constructs containing mutations that inhibit Rap1 binding, ICAP1α binding, disrupt KRIT1's phosphotyrosine-binding (PTB) domain, or direct KRIT1 to the plasma membrane, either alone or in combination, to restore barrier function in KRIT1-deficient endothelial cells. We found that ablating the 192NPAY195 motif or disrupting the PTB domain was sufficient to restore AJ protein localization and barrier function to control levels, irrespective of the junctional localization of KRIT1 or Rap1 binding. The ability of our KRIT1 constructs to rescue AJ and barrier function in KRIT1-depleted endothelial cells correlated with decreased ß1 integrin activity and maintenance of cortical actin fibers. Taken together, our findings indicate that Rap1 binding, ICAP1α binding and junctional localization are not required for the ability of KRIT1 to stabilize endothelial contacts, and suggest that the ability of KRIT1 to limit integrin activity could be involved in barrier stabilization.


Subject(s)
Endothelial Cells , Microtubule-Associated Proteins , Cell Communication , Integrin beta1 , KRIT1 Protein/genetics , Proto-Oncogene Proteins
5.
J Cell Sci ; 134(3)2021 02 04.
Article in English | MEDLINE | ID: mdl-33443102

ABSTRACT

KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion, and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC) in this process. In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol 12-myristate 13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein.


Subject(s)
Active Transport, Cell Nucleus , KRIT1 Protein/metabolism , Protein Kinase C-alpha , HeLa Cells , Humans , Phosphorylation , Protein Kinase C-alpha/genetics , Tetradecanoylphorbol Acetate
6.
Methods Mol Biol ; 2152: 259-265, 2020.
Article in English | MEDLINE | ID: mdl-32524558

ABSTRACT

Cerebral cavernous malformation (CCM) is driven by changes in the cerebral microvascular endothelial cell population. Mouse models of CCM have successfully recapitulated the disease in vivo; however, dissection of the disease pathogenesis and molecular mechanism is challenging in vivo due to limited access to the involved tissue in live animals. Therefore, in vitro tissue culture models are required. This protocol is designed to facilitate the isolation of cerebral microvascular endothelial cells from whole murine brain tissue. The protocol utilizes papain for a shorter, single digestion step to maximize cell recovery and viability. Using this technique, we are able to isolate cells from a murine CCM model in which the absence of CCM proteins is driven by Cre-mediated recombination at birth, and results in CCM-like vascular malformations in adult animals.


Subject(s)
Brain/metabolism , Cell Separation , Endothelial Cells/metabolism , KRIT1 Protein/genetics , Animals , Blood-Brain Barrier/metabolism , Cell Separation/methods , Disease Models, Animal , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Mutation
7.
Methods Mol Biol ; 2152: 387-400, 2020.
Article in English | MEDLINE | ID: mdl-32524567

ABSTRACT

Vascular permeability is a major function of the microvasculature that is regulated by multiple factors including blood pressure, blood viscosity, and endothelial barrier function. Intravital microscopy has been used to directly assess vascular permeability in vivo, which allows for the accurate measurement of endothelial barrier function in a truly physiological hemodynamic context. Here, we describe the procedure for measuring endothelial barrier function in mouse models of cerebral cavernous malformations, including micropipette preparation, anesthesia, tracheotomy, jugular catheterization, cremaster dissection, imaging, and data analysis. These animals exhibit an increase in microvessel permeability and abnormal vessel morphology, which require special consideration.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelium, Vascular/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/pathology , Intravital Microscopy , Animals , Blood-Brain Barrier/pathology , Data Analysis , Disease Models, Animal , Hemangioma, Cavernous, Central Nervous System/etiology , Intravital Microscopy/methods , Mice , Microvessels/metabolism , Microvessels/pathology
8.
Cell Mol Bioeng ; 13(2): 125-139, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32175026

ABSTRACT

INTRODUCTION: The pathophysiological increase in microvascular permeability plays a well-known role in the onset and progression of diseases like sepsis and atherosclerosis. However, how interactions between neutrophils and the endothelium alter vessel permeability is often debated. METHODS: In this study, we introduce a microfluidic, silicon-membrane enabled vascular mimetic (µSiM-MVM) for investigating the role of neutrophils in inflammation-associated microvascular permeability. In utilizing optically transparent silicon nanomembrane technology, we build on previous microvascular models by enabling in situ observations of neutrophil-endothelium interactions. To evaluate the effects of neutrophil transmigration on microvascular model permeability, we established and validated electrical (transendothelial electrical resistance and impedance) and small molecule permeability assays that allow for the in situ quantification of temporal changes in endothelium junctional integrity. RESULTS: Analysis of neutrophil-expressed ß1 integrins revealed a prominent role of neutrophil transmigration and basement membrane interactions in increased microvascular permeability. By utilizing blocking antibodies specific to the ß1 subunit, we found that the observed increase in microvascular permeability due to neutrophil transmigration is constrained when neutrophil-basement membrane interactions are blocked. Having demonstrated the value of in situ measurements of small molecule permeability, we then developed and validated a quantitative framework that can be used to interpret barrier permeability for comparisons to conventional Transwell™ values. CONCLUSIONS: Overall, our results demonstrate the potential of the µSiM-MVM in elucidating mechanisms involved in the pathogenesis of inflammatory disease, and provide evidence for a role for neutrophils in inflammation-associated endothelial barrier disruption.

9.
J Cell Mol Med ; 24(1): 632-639, 2020 01.
Article in English | MEDLINE | ID: mdl-31746130

ABSTRACT

The exact molecular mechanisms underlying CCM pathogenesis remain a complicated and controversial topic. Our previous work illustrated an important VEGF signalling loop in KRIT1 depleted endothelial cells. As VEGF is a major mediator of many vascular pathologies, we asked whether the increased VEGF signalling downstream of KRIT1 depletion was involved in CCM formation. Using an inducible KRIT1 endothelial-specific knockout mouse that models CCM, we show that VEGFR2 activation plays a role in CCM pathogenesis in mice. Inhibition of VEGFR2 using a specific inhibitor, SU5416, significantly decreased the number of lesions formed and slightly lowered the average lesion size. Notably, VEGFR2 inhibition also decreased the appearance of lesion haemorrhage as denoted by the presence of free iron in adjacent tissues. The presence of free iron correlated with increased microvessel permeability in both skeletal muscle and brain, which was completely reversed by SU5416 treatment. Finally, we show that VEGFR2 activation is a common downstream consequence of KRIT1, CCM2 and CCM3 loss of function, though the mechanism by which VEGFR2 activation occurs likely varies. Thus, our study clearly shows that VEGFR2 activation downstream of KRIT1 depletion enhances the severity of CCM formation in mice, and suggests that targeting VEGF signalling may be a potential future therapy for CCM.


Subject(s)
Endothelial Cells/pathology , Hemangioma, Cavernous, Central Nervous System/pathology , Hemorrhagic Stroke/pathology , KRIT1 Protein/physiology , Pulmonary Artery/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemorrhagic Stroke/etiology , Hemorrhagic Stroke/metabolism , Male , Mice , Mice, Knockout , Pulmonary Artery/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
10.
Sci Rep ; 7(1): 8296, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811547

ABSTRACT

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.


Subject(s)
Endothelium/metabolism , KRIT1 Protein/deficiency , NADPH Oxidases/metabolism , Oxidation-Reduction , Signal Transduction , Animals , Antioxidants/metabolism , Capillary Permeability/drug effects , Capillary Permeability/genetics , Gene Expression Regulation , KRIT1 Protein/genetics , KRIT1 Protein/metabolism , Mice , Mice, Knockout , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS One ; 11(9): e0162338, 2016.
Article in English | MEDLINE | ID: mdl-27612188

ABSTRACT

The phosphoinositide-specific phospholipase C, PLCε, is a unique signaling protein with known roles in regulating cardiac myocyte growth, astrocyte inflammatory signaling, and tumor formation. PLCε is also expressed in endothelial cells, however its role in endothelial regulation is not fully established. We show that endothelial cells of multiple origins, including human pulmonary artery (HPAEC), human umbilical vein (HUVEC), and immortalized brain microvascular (hCMEC/D3) endothelial cells, express PLCε. Knockdown of PLCε in arterial endothelial monolayers decreased the effectiveness of the endothelial barrier. Concomitantly, RhoA activity and stress fiber formation were increased. PLCε-deficient arterial endothelial cells also exhibited decreased Rap1-GTP levels, which could be restored by activation of the Rap1 GEF, Epac, to rescue the increase in monolayer leak. Reintroduction of PLCε rescued monolayer leak with both the CDC25 GEF domain and the lipase domain of PLCε required to fully activate Rap1 and to rescue endothelial barrier function. Finally, we demonstrate that the barrier promoting effects PLCε are dependent on Rap1 signaling through the Rap1 effector, KRIT1, which we have previously shown is vital for maintaining endothelial barrier stability. Thus we have described a novel role for PLCε PIP2 hydrolytic and Rap GEF activities in arterial endothelial cells, where PLCε-dependent activation of Rap1/KRIT1 signaling promotes endothelial barrier stability.


Subject(s)
Endothelial Cells/metabolism , Phosphoinositide Phospholipase C/metabolism , Animals , Cell Line , Endothelium, Vascular/physiology , Gene Knockdown Techniques , Humans , KRIT1 Protein , Lipase/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoinositide Phospholipase C/genetics , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , cdc25 Phosphatases/metabolism , rhoA GTP-Binding Protein/metabolism
12.
Int J Biochem Cell Biol ; 81(Pt B): 254-270, 2016 12.
Article in English | MEDLINE | ID: mdl-27639680

ABSTRACT

Cerebral Cavernous Malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or is inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions exhibit a range of different phenotypes, including wide inter-individual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Lesions may remain asymptomatic or result in pathological conditions of various type and severity at any age, with symptoms ranging from recurrent headaches to severe neurological deficits, seizures, and stroke. To date there are no direct therapeutic approaches for CCM disease besides the surgical removal of accessible lesions. Novel pharmacological strategies are particularly needed to limit disease progression and severity and prevent de novo formation of CCM lesions in susceptible individuals. Useful insights into innovative approaches for CCM disease prevention and treatment are emerging from a growing understanding of the biological functions of the three known CCM proteins, CCM1/KRIT1, CCM2 and CCM3/PDCD10. In particular, accumulating evidence indicates that these proteins play major roles in distinct signaling pathways, including those involved in cellular responses to oxidative stress, inflammation and angiogenesis, pointing to pathophysiological mechanisms whereby the function of CCM proteins may be relevant in preventing vascular dysfunctions triggered by these events. Indeed, emerging findings demonstrate that the pleiotropic roles of CCM proteins reflect their critical capacity to modulate the fine-tuned crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses, providing a novel mechanistic scenario that reconciles both the multiple signaling pathways linked to CCM proteins and the distinct therapeutic approaches proposed so far. In addition, recent studies in CCM patient cohorts suggest that genetic susceptibility factors related to differences in vascular sensitivity to oxidative stress and inflammation contribute to inter-individual differences in CCM disease susceptibility and severity. This review discusses recent progress into the understanding of the molecular basis and mechanisms of CCM disease pathogenesis, with specific emphasis on the potential contribution of altered cell responses to oxidative stress and inflammatory events occurring locally in the microvascular environment, and consequent implications for the development of novel, safe, and effective preventive and therapeutic strategies.


Subject(s)
Hemangioma, Cavernous, Central Nervous System/physiopathology , Inflammation , Oxidative Stress , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology
13.
Tissue Barriers ; 3(1-2): e985954, 2015.
Article in English | MEDLINE | ID: mdl-25838987

ABSTRACT

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.

14.
J Biol Chem ; 289(47): 33054-65, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25320085

ABSTRACT

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.


Subject(s)
Endothelial Cells/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Capillary Permeability , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cattle , Cell Communication , Cell Membrane Permeability , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , KRIT1 Protein , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , RNA Interference , Vascular Endothelial Growth Factor Receptor-2/metabolism , beta Catenin/metabolism
15.
Arterioscler Thromb Vasc Biol ; 32(11): 2702-10, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22922958

ABSTRACT

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.


Subject(s)
Arterioles/metabolism , Arthritis/metabolism , Capillary Permeability , Edema/metabolism , Microtubule-Associated Proteins/deficiency , Proto-Oncogene Proteins/deficiency , Venules/metabolism , Animals , Arterioles/immunology , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Down-Regulation , Edema/genetics , Edema/immunology , Edema/pathology , Homeostasis , Humans , Inflammation Mediators/metabolism , Joints/immunology , Joints/metabolism , Joints/pathology , KRIT1 Protein , Leukocyte Rolling , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Venules/immunology
16.
Dis Model Mech ; 3(1-2): 73-83, 2010.
Article in English | MEDLINE | ID: mdl-20007487

ABSTRACT

KRIT1, also called CCM1, is a member of a multiprotein complex that contains the products of the CCM2 and PDCD10 (also known as CCM3) loci. Heterozygous loss of any of the genes that encode these proteins leads to cerebral cavernous malformations (CCM), which are vascular lesions that are found in around 0.5% of humans. KRIT1 mediates the stabilization of beta-catenin-containing endothelial cell-cell junctions downstream of the Rap1 GTPase. Here, we report that Rap1 and KRIT1 are negative regulators of canonical beta-catenin signaling in mice and that hemizygous Krit1 deficiency exacerbates beta-catenin-driven pathologies. Depletion of endothelial KRIT1 caused beta-catenin to dissociate from vascular endothelial (VE)-cadherin and to accumulate in the nucleus with consequent increases in beta-catenin-dependent transcription. Activation of Rap1 inhibited beta-catenin-dependent transcription in confluent endothelial cells; this effect required the presence of intact cell-cell junctions and KRIT1. These effects of KRIT1 were not limited to endothelial cells; the KRIT1 protein was expressed widely and its depletion increased beta-catenin signaling in epithelial cells. Moreover, a reduction in KRIT1 expression also increased beta-catenin signaling in vivo. Hemizygous deficiency of Krit1 resulted in a ~1.5-fold increase in intestinal polyps in the Apc(Min/+) mouse, which was associated with increased beta-catenin-driven transcription. Thus, KRIT1 regulates beta-catenin signaling, and Krit1(+/-) mice are more susceptible to beta-catenin-driven intestinal adenomas.


Subject(s)
Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , beta Catenin/metabolism , rap1 GTP-Binding Proteins/metabolism , Adenoma/genetics , Adenoma/pathology , Animals , Cadherins/metabolism , Cattle , Cell Nucleus/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , KRIT1 Protein , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Organ Specificity , Protein Transport , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Survival Analysis , Transcription, Genetic
17.
J Cell Biol ; 179(2): 247-54, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17954608

ABSTRACT

Cerebral cavernous malformation (CCM), a disease associated with defective endothelial junctions, result from autosomal dominant CCM1 mutations that cause loss of KRIT-1 protein function, though how the loss of KRIT-1 leads to CCM is obscure. KRIT-1 binds to Rap1, a guanosine triphosphatase that maintains the integrity of endothelial junctions. Here, we report that KRIT-1 protein is expressed in cultured arterial and venous endothelial cells and is present in cell-cell junctions. KRIT-1 colocalized and was physically associated with junctional proteins via its band 4.1/ezrin/radixin/moesin (FERM) domain. Rap1 activity regulated the junctional localization of KRIT-1 and its physical association with junction proteins. However, the association of the isolated KRIT-1 FERM domain was independent of Rap1. Small interfering RNA-mediated depletion of KRIT-1 blocked the ability of Rap1 to stabilize endothelial junctions associated with increased actin stress fibers. Thus, Rap1 increases KRIT-1 targeting to endothelial cell-cell junctions where it suppresses stress fibers and stabilizes junctional integrity.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CHO Cells , Cattle , Cell Membrane Permeability/drug effects , Cricetinae , Cricetulus , Humans , KRIT1 Protein , Microtubule-Associated Proteins/chemistry , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Proto-Oncogene Proteins/chemistry
18.
Cancer Res ; 67(4): 1536-44, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308092

ABSTRACT

Phosphoprotein enriched in astrocytes of 15 kDa (PEA-15) binds to extracellular signal-regulated kinase 1 and 2 (ERK1/2) mitogen-activated protein (MAP) kinases to alter ERK1/2 cellular localization and target preferences and binds to adaptors in the extrinsic cell death pathway to block apoptosis. Here, we report that PEA-15 protein expression is inversely correlated with the invasive behavior of breast cancer in an immunohistochemical analysis of a breast cancer progression tissue microarray. Short hairpin RNA-mediated inhibition of PEA-15 expression increased the invasion of PEA-15-expressing tumor cells in vitro, suggesting a causative role for PEA-15 in the inhibition of invasion. This causative role was confirmed by the finding that the enforced expression of PEA-15 in invasive tumor cells reduced invasion. The effect of PEA-15 on tumor invasion is mediated by its interaction with ERK1/2 as shown by the following: (a) PEA-15 mutants that fail to bind ERK1/2 did not inhibit invasion; (b) overexpression of ERK1 or activated MAP/ERK kinase (MEK) reversed the inhibitory effect of PEA-15; (c) when an inhibitor of ERK1/2 activation reduced invasion, PEA-15 expression did not significantly reduce invasion further. Furthermore, we find that the effect of PEA-15 on invasion seems to relate to the nuclear localization of activated ERK1/2. PEA-15 inhibits invasion by keeping ERK out of the nucleus, as a PEA-15 mutant that cannot prevent ERK nuclear localization was not able to inhibit invasion. In addition, membrane-localized ERK1, which sequesters endogenous ERK1 to prevent its nuclear localization, also inhibited invasion. These results reveal that PEA-15 regulates cancer cell invasion via its ability to bind ERK1/2 and indicate that nuclear entry of ERK1/2 is important in tumor behavior.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Animals , Apoptosis Regulatory Proteins , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Line, Tumor , Cell Nucleus/enzymology , Cricetinae , Cricetulus , Glioblastoma/enzymology , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/genetics , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Protein Binding , Transfection
19.
Mol Biol Cell ; 16(8): 3552-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15917297

ABSTRACT

Cell cycle progression is dependent on the nuclear localization and transcriptional effects of activated extracellular signal-regulated kinase (ERK)1 and ERK2 mitogen-activated protein (MAP) kinases (ERK1/2). Phosphoprotein enriched in astrocytes (PEA-15) binds ERK1/2 and inhibits their nuclear localization, thus blocking cell proliferation. Here, we report that phosphorylation of PEA-15 blocks its interaction with ERK1/2 in vitro and in vivo and that phosphorylation of both Ser104 and Ser116 is required for this effect. Using phosphomimetic and nonphosphorylatable mutants of PEA-15, we found that PEA-15 phosphorylation abrogates its capacity to block the nuclear localization and transcriptional activities of ERK1/2; this phosphorylation therefore enables the proliferation of cells that express high levels of PEA-15. Additionally, we report that PEA-15 phosphorylation can modulate nontranscriptional activities of ERK1/2, such as the modulation of the affinity of integrin adhesion receptors. Finally, we used a novel anti-phospho-specific PEA-15 antibody to establish that PEA-15 is phosphorylated in situ in normal mammary epithelium. These results define a novel posttranslational mechanism for controlling the subcellular localization of ERK1/2 and for specifying the output of MAP kinase signaling.


Subject(s)
Astrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Phosphoproteins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Proliferation , Cricetinae , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation/genetics , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Transcription, Genetic
20.
Mol Cell Biol ; 25(5): 1922-41, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15713646

ABSTRACT

Keratinocyte migration is critical to reepithelialization during wound repair. The motility response is promoted by growth factors, cytokines, and cytokines produced in the wound bed, including those that activate the epidermal growth factor (EGF) receptor. The Alu-Leu-Arg-negative CXC chemokine interferon-inducible protein 9 (IP-9; also known as CXCL11, I-TAC, beta-R1, and H-174) is produced by keratinocytes in response to injury. As keratinocytes also express the receptor, CXCR3, this prompted us to examine the role and molecular mechanism by which IP-9 regulates keratinocyte motility. Unexpectedly, as CXCR3 liganding blocks growth factor-induced motility in fibroblasts, IP-9 alone promoted motility in undifferentiated keratinocytes (37 +/- 6% of the level of the highly motogenic EGF) as determined in a two-dimensional in vitro wound healing assay. IP-9 even enhanced EGF-induced motility in undifferentiated keratinocytes (116 +/- 5%; P < 0.05 compared to EGF alone), suggesting two separate mechanisms of action. IP-9-increased motility and -decreased adhesiveness required the intracellular protease calpain. The increases in both motility and calpain activity by IP-9 were blocked by pharmacological and molecular inhibition of phospholipase C-beta3 and chelation of calcium, which prevented an intracellular calcium flux. Molecular downregulation or RNA interference-mediated depletion of mu-calpain (calpain 1) but not M-calpain (calpain 2) blocked IP-9-induced calpain activation and motility. In accord with elimination of IP-9-induced de-adhesion, RNA interference-mediated depletion of calpain 1 but not calpain 2 prevented cleavage of the focal adhesion component focal adhesion kinase and disassembly of vinculin aggregates. In comparison, EGF-induced motility of the same undifferentiated keratinocytes requires the previously described extracellular signal-regulated kinase to the M-calpain pathway. These data demonstrate that while both EGF- and IP-9-induced motility in keratinocytes requires calpain activity, the isoform of calpain triggered depends on the nature of the receptor for the particular ligand. Interestingly, physiological nonapoptotic calcium fluxes were capable of activating mu-calpain, implying that the calcium requirement of mu-calpain for activation is attained during cell signaling. This is also the first demonstration of differential activation of the two ubiquitous calpain isoforms in the same cell by different signals.


Subject(s)
Calcium/metabolism , Calpain/metabolism , Cell Movement/physiology , Chemokines, CXC/physiology , Egtazic Acid/analogs & derivatives , Keratinocytes/physiology , Calpain/genetics , Cell Line , Chelating Agents/pharmacology , Chemokine CXCL11 , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Down-Regulation , Egtazic Acid/pharmacology , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Interferons/physiology , Keratinocytes/chemistry , Keratinocytes/drug effects , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Vinculin/analysis , Vinculin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...