Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biochem Soc Trans ; 52(1): 137-150, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38323651

ABSTRACT

Neuropilin-1 (NRP1) is a transmembrane glycoprotein expressed by several cell types including, neurons, endothelial cells (ECs), smooth muscle cells, cardiomyocytes and immune cells comprising macrophages, dendritic cells and T cell subsets. Since NRP1 discovery in 1987 as an adhesion molecule in the frog nervous system, more than 2300 publications on PubMed investigated the function of NRP1 in physiological and pathological contexts. NRP1 has been characterised as a coreceptor for class 3 semaphorins and several members of the vascular endothelial growth factor (VEGF) family. Because the VEGF family is the main regulator of blood and lymphatic vessel growth in addition to promoting neurogenesis, neuronal patterning, neuroprotection and glial growth, the role of NRP1 in these biological processes has been extensively investigated. It is now established that NRP1 promotes the physiological growth of new vessels from pre-existing ones in the process of angiogenesis. Furthermore, several studies have shown that NRP1 mediates signalling pathways regulating pathological vascular growth in ocular neovascular diseases and tumour development. Less defined are the roles of NRP1 in maintaining the function of the quiescent established vasculature in an adult organism. This review will focus on the opposite roles of NRP1 in regulating transforming growth factor ß signalling pathways in different cell types, and on the emerging role of endothelial NRP1 as an atheroprotective, anti-inflammatory factor involved in the response of ECs to shear stress.


Subject(s)
Atherosclerosis , Neuropilin-1 , Humans , Adult , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Angiogenesis , Inflammation
2.
Sci Signal ; 16(786): eabo4863, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37220183

ABSTRACT

Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-ß (TGF-ß) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-ß signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-ß signaling and inflammation.


Subject(s)
Endothelial Cells , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type II , Animals , Mice , Adherens Junctions , Endothelium , Cadherins
4.
Cells ; 9(9)2020 09 08.
Article in English | MEDLINE | ID: mdl-32911833

ABSTRACT

Endothelial cells drive the formation of new blood vessels in physiological and pathological contexts such as embryonic development, wound healing, cancer and ocular diseases. Once formed, all vessels of the vasculature system present an endothelial monolayer (the endothelium), lining the luminal wall of the vessels, that regulates gas and nutrient exchange between the circulating blood and tissues, contributing to maintaining tissue and vascular homeostasis. To perform their functions, endothelial cells integrate signalling pathways promoted by growth factors, cytokines, extracellular matrix components and signals from mechanosensory complexes sensing the blood flow. New evidence shows that endothelial cells rely on specific metabolic pathways for distinct cellular functions and that the integration of signalling and metabolic pathways regulates endothelial-dependent processes such as angiogenesis and vascular homeostasis. In this review, we provide an overview of endothelial functions and the recent advances in understanding the role of endothelial signalling and metabolism in physiological processes such as angiogenesis and vascular homeostasis and vascular diseases. Also, we focus on the signalling pathways promoted by the transmembrane protein Neuropilin-1 (NRP1) in endothelial cells, its recently discovered role in regulating mitochondrial function and iron homeostasis and the role of mitochondrial dysfunction and iron in atherosclerosis and neurodegenerative diseases.


Subject(s)
Alzheimer Disease/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Iron/metabolism , Homeostasis , Humans , Metabolic Networks and Pathways , Reactive Oxygen Species , Signal Transduction
6.
iScience ; 11: 205-223, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30623799

ABSTRACT

The transmembrane protein neuropilin-1 (NRP1) promotes vascular endothelial growth factor (VEGF) and extracellular matrix signaling in endothelial cells (ECs). Although it is established that NRP1 is essential for angiogenesis, little is known about its role in EC homeostasis. Here, we report that NRP1 promotes mitochondrial function in ECs by preventing iron accumulation and iron-induced oxidative stress through a VEGF-independent mechanism in non-angiogenic ECs. Furthermore, NRP1-deficient ECs have reduced growth and show the hallmarks of cellular senescence. We show that a subcellular pool of NRP1 localizes in mitochondria and interacts with the mitochondrial transporter ATP-binding cassette B8 (ABCB8). NRP1 loss reduces ABCB8 levels, resulting in iron accumulation, iron-induced mitochondrial superoxide production, and iron-dependent EC senescence. Treatment of NRP1-deficient ECs with the mitochondria-targeted antioxidant compound mitoTEMPO or with the iron chelator deferoxamine restores mitochondrial activity, inhibits superoxide production, and protects from cellular senescence. This finding identifies an unexpected role of NRP1 in EC homeostasis.

7.
Nat Commun ; 8(1): 895, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026072

ABSTRACT

The role of the endothelium in protecting from chronic liver disease and TGFß-mediated fibrosis remains unclear. Here we describe how the endothelial transcription factor ETS-related gene (ERG) promotes liver homoeostasis by controlling canonical TGFß-SMAD signalling, driving the SMAD1 pathway while repressing SMAD3 activity. Molecular analysis shows that ERG binds to SMAD3, restricting its access to DNA. Ablation of ERG expression results in endothelial-to-mesenchymal transition (EndMT) and spontaneous liver fibrogenesis in EC-specific constitutive hemi-deficient (Erg cEC-Het ) and inducible homozygous deficient mice (Erg iEC-KO ), in a SMAD3-dependent manner. Acute administration of the TNF-α inhibitor etanercept inhibits carbon tetrachloride (CCL4)-induced fibrogenesis in an ERG-dependent manner in mice. Decreased ERG expression also correlates with EndMT in tissues from patients with end-stage liver fibrosis. These studies identify a pathogenic mechanism where loss of ERG causes endothelial-dependent liver fibrogenesis via regulation of SMAD2/3. Moreover, ERG represents a promising candidate biomarker for assessing EndMT in liver disease.The transcription factor ERG is key to endothelial lineage specification and vascular homeostasis. Here the authors show that ERG balances TGFß signalling through the SMAD1 and SMAD3 pathways, protecting the endothelium from endothelial-to-mesenchymal transition and consequent liver fibrosis in mice via a SMAD3-dependent mechanism.


Subject(s)
Endothelial Cells/metabolism , Liver Cirrhosis, Biliary/pathology , Liver/pathology , Oncogene Proteins/metabolism , Transcriptional Regulator ERG/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carbon Tetrachloride/toxicity , Cells, Cultured , Down-Regulation , End Stage Liver Disease/etiology , End Stage Liver Disease/surgery , Epithelial-Mesenchymal Transition , Etanercept/pharmacology , Etanercept/therapeutic use , Female , Fibrosis , Human Umbilical Vein Endothelial Cells , Humans , Liver/drug effects , Liver/surgery , Liver Cirrhosis, Biliary/complications , Liver Cirrhosis, Biliary/therapy , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/genetics , Signal Transduction/drug effects , Smad1 Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Regulator ERG/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation
8.
J Exp Med ; 214(4): 1049-1064, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28289053

ABSTRACT

The vascular endothelial growth factor (VEGF) isoform VEGF165 stimulates vascular growth and hyperpermeability. Whereas blood vessel growth is essential to sustain organ health, chronic hyperpermeability causes damaging tissue edema. By combining in vivo and tissue culture models, we show here that VEGF165-induced vascular leakage requires both VEGFR2 and NRP1, including the VEGF164-binding site of NRP1 and the NRP1 cytoplasmic domain (NCD), but not the known NCD interactor GIPC1. In the VEGF165-bound receptor complex, the NCD promotes ABL kinase activation, which in turn is required to activate VEGFR2-recruited SRC family kinases (SFKs). These results elucidate the receptor complex and signaling hierarchy of downstream kinases that transduce the permeability response to VEGF165. In a mouse model with choroidal neovascularisation akin to age-related macular degeneration, NCD loss attenuated vessel leakage without affecting neovascularisation. These findings raise the possibility that targeting NRP1 or its NCD interactors may be a useful therapeutic strategy in neovascular disease to reduce VEGF165-induced edema without compromising vessel growth.


Subject(s)
Capillary Permeability , Neuropilin-1/physiology , Proto-Oncogene Proteins c-abl/physiology , Vascular Endothelial Growth Factor A/physiology , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/physiology , Animals , Enzyme Activation , Mice , Mice, Inbred C57BL , Semaphorin-3A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
10.
Sci Rep ; 6: 26142, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27199173

ABSTRACT

Strong evidence suggests that phospholipase Cγ1 (PLCγ1) is a suitable target to counteract tumourigenesis and metastasis dissemination. We recently identified a novel signalling pathway required for PLCγ1 activation which involves formation of a protein complex with 3-phosphoinositide-dependent protein kinase 1 (PDK1). In an effort to define novel strategies to inhibit PLCγ1-dependent signals we tested here whether a newly identified and highly specific PDK1 inhibitor, 2-O-benzyl-myo-inositol 1,3,4,5,6-pentakisphosphate (2-O-Bn-InsP5), could affect PDK1/PLCγ1 interaction and impair PLCγ1-dependent cellular functions in cancer cells. Here, we demonstrate that 2-O-Bn-InsP5 interacts specifically with the pleckstrin homology domain of PDK1 and impairs formation of a PDK1/PLCγ1 complex. 2-O-Bn-InsP5 is able to inhibit the epidermal growth factor-induced PLCγ1 phosphorylation and activity, ultimately resulting in impaired cancer cell migration and invasion. Importantly, we report that 2-O-Bn-InsP5 inhibits cancer cell dissemination in zebrafish xenotransplants. This work demonstrates that the PDK1/PLCγ1 complex is a potential therapeutic target to prevent metastasis and it identifies 2-O-Bn-InsP5 as a leading compound for development of anti-metastatic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Inositol Phosphates/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Heterografts , Humans , Melanoma/drug therapy , Neoplasm Transplantation , Protein Binding , Protein Multimerization , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Zebrafish
11.
Prog Retin Eye Res ; 52: 64-83, 2016 05.
Article in English | MEDLINE | ID: mdl-26923176

ABSTRACT

Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.


Subject(s)
Eye Diseases , Neovascularization, Pathologic , Neuropilin-1/metabolism , Animals , Brain/cytology , Endothelium, Vascular/metabolism , Eye Diseases/genetics , Eye Diseases/metabolism , Eye Diseases/therapy , Humans , Models, Molecular , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Neurons/metabolism , Neuropilin-1/genetics , Retina/cytology , Signal Transduction/physiology
12.
Cell Rep ; 11(10): 1577-90, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26051942

ABSTRACT

Sprouting blood vessels are led by filopodia-studded endothelial tip cells that respond to angiogenic signals. Mosaic lineage tracing previously revealed that NRP1 is essential for tip cell function, although its mechanistic role in tip cells remains poorly defined. Here, we show that NRP1 is dispensable for genetic tip cell identity. Instead, we find that NRP1 is essential to form the filopodial bursts that distinguish tip cells morphologically from neighboring stalk cells, because it enables the extracellular matrix (ECM)-induced activation of CDC42, a key regulator of filopodia formation. Accordingly, NRP1 knockdown and pharmacological CDC42 inhibition similarly impaired filopodia formation in vitro and in developing zebrafish in vivo. During mouse retinal angiogenesis, CDC42 inhibition impaired tip cell and vascular network formation, causing defects that resembled those due to loss of ECM-induced, but not VEGF-induced, NRP1 signaling. We conclude that NRP1 enables ECM-induced filopodia formation for tip cell function during sprouting angiogenesis.


Subject(s)
Endothelial Cells/cytology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Pseudopodia/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , Animals , Embryo, Mammalian , Endothelial Cells/metabolism , Immunohistochemistry , Mice , Neovascularization, Physiologic/physiology , Rhombencephalon/blood supply , Rhombencephalon/cytology , Signal Transduction , Zebrafish
13.
Mol Cell Oncol ; 2(1): e968034, 2015.
Article in English | MEDLINE | ID: mdl-27308396

ABSTRACT

We recently reported that neuropilin 1 (NRP1) drives angiogenesis by promoting extracellular matrix signaling in endothelial cells via ABL1 kinase. Imatinib targets this pathway in pathological angiogenesis and may provide a novel opportunity for anti-angiogenic therapy of age-related macular degeneration, proliferative diabetic retinopathy, or solid tumor growth.

14.
Biochem Soc Trans ; 42(5): 1429-34, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25233427

ABSTRACT

Neuropilin-1 (NRP1), together with neuropilin-2, belongs to the neuropilin family. Neuropilins are transmembrane proteins essential for vascular and neural development and act as co-receptors for secreted signalling molecules of the class 3 semaphorin and vascular endothelial growth factor A (VEGF-A) families. NRP1 promotes VEGF-A signal in blood vascular endothelium and semaphorin signal in lymphatic endothelium, by forming complexes with its co-receptors. Mouse mutant studies established that NRP1 expression is essential during development because mice lacking NRP1 expression die embryonically and show severe neuronal and cardiovascular defects. Even though the contribution of NRP1 to vascular development has been mainly ascribed to its function as a VEGF-A receptor, recent evidence suggests that NRP1 contributes to angiogenesis through VEGF-independent mechanisms. In the present paper, we provide an overview of NRP1 functions in the vasculature and discuss current knowledge of NRP1-dependent signalling in the endothelium.


Subject(s)
Endothelium, Vascular/metabolism , Extracellular Matrix , Models, Biological , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Neuropilin-1/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Humans , Mice, Mutant Strains , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/agonists , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
J Exp Med ; 211(6): 1167-83, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24863063

ABSTRACT

To enable new blood vessel growth, endothelial cells (ECs) express neuropilin 1 (NRP1), and NRP1 associates with the receptor tyrosine kinase VEGFR2 after binding the vascular endothelial growth factor A (VEGF) to enhance arteriogenesis. We report that NRP1 contributes to angiogenesis through a novel mechanism. In human and mouse ECs, the integrin ligand fibronectin (FN) stimulated actin remodeling and phosphorylation of the focal adhesion component paxillin (PXN) in a VEGF/VEGFR2-independent but NRP1-dependent manner. NRP1 formed a complex with ABL1 that was responsible for FN-dependent PXN activation and actin remodeling. This complex promoted EC motility in vitro and during angiogenesis on FN substrates in vivo. Accordingly, both physiological and pathological angiogenesis in the retina were inhibited by treatment with Imatinib, a small molecule inhibitor of ABL1 which is widely used to prevent the proliferation of tumor cells that express BCR-ABL fusion proteins. The finding that NRP1 regulates angiogenesis in a VEGF- and VEGFR2-independent fashion via ABL1 suggests that ABL1 inhibition provides a novel opportunity for anti-angiogenic therapy to complement VEGF or VEGFR2 blockade in eye disease or solid tumor growth.


Subject(s)
Benzamides/pharmacology , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Neuropilin-1/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Animals , Cell Adhesion/genetics , Cell Movement/genetics , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Enzyme Activation/drug effects , Fibronectins/metabolism , Humans , Imatinib Mesylate , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Neuropilin-1/genetics , Paxillin/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/genetics , RNA Interference , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
J Cell Sci ; 127(Pt 14): 3079-93, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24777476

ABSTRACT

The protein iASPP (encoded by PPP1R13L) is an evolutionarily conserved p53 inhibitor, the expression of which is often upregulated in human cancers. We have recently shown that iASPP is a crucial regulator of epidermal homeostasis. Here, we report that iASPP also acts as autophagy inhibitor in keratinocytes. Our data show that depletion of iASPP protects keratinocytes from apoptosis by modulating the expression of Noxa (also known as PMAIP1). In our model, iASPP expression can affect the fission-fusion cycle, mass and shape of mitochondria. iASPP-silenced keratinocytes display disorganization of cytosolic compartments and increased metabolic stress caused by deregulation of mTORC1 signaling. Moreover, increased levels of lipidated LC3 protein confirmed the activation of autophagy in iASPP-depleted cells. We have identified a novel mechanism modulating autophagy in keratinocytes that relies upon iASPP expression specifically reducing the interaction of Atg5-Atg12 with Atg16L1, an interaction that is essential for autophagosome formation or maturation. Using organotypic culture, we further explored the link between autophagy and differentiation, and we showed that impairing autophagy affects epidermal terminal differentiation. Our data provide an alternative mechanism to explain how epithelial integrity is maintained against environmental stressors and might also improve the understanding of the etiology of skin diseases that are characterized by defects in differentiation and DNA damage responses.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Apoptosis/physiology , Autophagy/physiology , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Carrier Proteins/metabolism , Cell Differentiation/physiology , Epidermal Cells , Epidermis/metabolism , Humans , Microtubule-Associated Proteins/metabolism
17.
Semin Cell Dev Biol ; 24(3): 172-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319134

ABSTRACT

The neuropilins NRP1 and NRP2 are transmembrane proteins that regulate many different aspects of vascular and neural development. Even though they were originally identified as adhesion molecules, they are most commonly studied as co-receptors for secreted signalling molecules of the class 3 semaphorin (SEMA) and vascular endothelial growth factor (VEGF) families. During nervous system development, both classes of ligands control soma migration, axon patterning and synaptogenesis in the central nervous system, and they additionally help to guide the neural crest cell precursors of neurons and glia in the peripheral nervous system. Both classes of neuropilin ligands also control endothelial cell behaviour, with NRP1 acting as a VEGF-A isoform receptor in blood vascular endothelium and as a semaphorin receptor in lymphatic valve endothelium, and NRP2 promoting lymphatic vessel growth induced by VEGF-C. Here we provide an overview of neuropilin function in neurons and neural crest cells, discuss current knowledge of neuropilin signalling in the vasculature and conclude with a summary of neuropilin roles in cancer.


Subject(s)
Neoplasms/metabolism , Neurons/metabolism , Neuropilins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Humans , Neoplasms/blood supply , Semaphorins/metabolism
18.
J Cell Sci ; 125(Pt 13): 3153-63, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22454520

ABSTRACT

3-Phosphoinositide-dependent protein kinase-1 (PDK1) and phospholipase C (PLC)γ1 are two key enzymes in signal transduction that control several intracellular processes. Despite the fact that PLCγ1 has been investigated for several years, the mechanisms of activation of this enzyme are still not completely clear. Similarly, although PDK1 has been mostly investigated for its role in activation of Akt, a crucial enzyme in regulation of several cellular processes, it has become evident recently that the role of PDK1 in physiological and pathological conditions is not limited to Akt activation. Here we demonstrate that PDK1 regulates PLCγ1 activation in a mechanism involving association of the two enzymes and modulation of PLCγ1 tyrosine phosphorylation. We further show that this novel PDK1-PLCγ1 pathway is important for cancer cell invasion. The identification of a PDK1-PLCγ1 pathway reveals the existence of a previously undetected link between two of the most important enzymes in signal transduction. This is likely to have profound consequences for our understanding of several cellular functions that are dependent on phosphoinositides and controlled by PDK1 and PLCγ1.


Subject(s)
Gene Expression Regulation, Neoplastic , Phospholipase C gamma/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases , Calcium/metabolism , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Flow Cytometry , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Indazoles/pharmacology , Neoplasm Invasiveness/genetics , Phospholipase C gamma/genetics , Phosphorylation , Protein Interaction Mapping , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
20.
EMBO J ; 30(20): 4261-73, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21897369

ABSTRACT

iASPP, an inhibitory member of the ASPP (apoptosis stimulating protein of p53) family, is an evolutionarily conserved inhibitor of p53 which is frequently upregulated in human cancers. However, little is known about the role of iASPP under physiological conditions. Here, we report that iASPP is a critical regulator of epithelial development. We demonstrate a novel autoregulatory feedback loop which controls crucial physiological activities by linking iASPP to p63, via two previously unreported microRNAs, miR-574-3p and miR-720. By investigating its function in stratified epithelia, we show that iASPP participates in the p63-mediated epithelial integrity program by regulating the expression of genes essential for cell adhesion. Silencing of iASPP in keratinocytes by RNA interference promotes and accelerates a differentiation pathway, which also affects and slowdown cellular proliferation. Taken together, these data reveal iASPP as a key regulator of epithelial homeostasis.


Subject(s)
Feedback, Physiological , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Adhesion , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Gene Expression , HEK293 Cells , Humans , Keratinocytes/metabolism , Mice , MicroRNAs/metabolism , RNA Interference , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL