Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
FASEB J ; 35(6): e21627, 2021 06.
Article in English | MEDLINE | ID: mdl-33948992

ABSTRACT

Capillary endothelial cells (ECs) maintain a semi-permeable barrier between the blood and tissue by forming inter-EC tight junctions (TJs), regulating selective transport of fluid and solutes. Overwhelming inflammation, as occurs in sepsis, disrupts these TJs, leading to leakage of fluid, proteins, and small molecules into the tissues. Mechanistically, disruption of capillary barrier function is mediated by small Rho-GTPases, such as RhoA, -B, and -C, which are activated by guanine nucleotide exchange factors (GEFs) and disrupted by GTPase-activating factors (GAPs). We previously reported that a mutation in a specific RhoB GAP (p190BRhoGAP) underlays a hereditary capillary leak syndrome. Tumor necrosis factor (TNF) treatment disrupts TJs in cultured human microvascular ECs, a model of capillary leak. This response requires new gene transcription and involves increased RhoB activation. However, the specific GEF that activates RhoB in capillary ECs remains unknown. Transcriptional profiling of cultured tight junction-forming human dermal microvascular endothelial cells (HDMECs) revealed that 17 GEFs were significantly induced by TNF. The function of each candidate GEF was assessed by short interfering RNA depletion and trans-endothelial electrical resistance screening. Knockown of ArhGEF10 reduced the TNF-induced loss of barrier which was phenocopied by RhoB or dual ArhGEF10/RhoB knockdown. ArhGEF10 knockdown also reduced the extent of TNF-induced RhoB activation and disruption at tight junctions. In a cell-free assay, immunoisolated ArhGEF10 selectively catalyzed nucleotide exchange to activate RhoB, but not RhoA or RhoC. We conclude ArhGEF10 is a TNF-induced RhoB-selective GEF that mediates TJ disruption and barrier loss in human capillary endothelial cells.


Subject(s)
Dermis/metabolism , Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Tight Junctions/physiology , rhoB GTP-Binding Protein/metabolism , Capillary Permeability , Dermis/cytology , Dermis/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , rhoB GTP-Binding Protein/genetics
2.
J Exp Med ; 214(12): 3497-3505, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29097442

ABSTRACT

We describe a fatal case of pediatric systemic capillary leak (Clarkson's disease) associated with a point mutation in p190BRhoGAP. Dermal microvascular endothelial cells (ECs) isolated from this patient form monolayers with similar levels and distribution of junctional proteins and transendothelial electrical resistance compared with normal human dermal microvascular ECs. However, patient-derived ECs demonstrate a greater increase in permeability and impaired recovery of barrier function in response to tumor necrosis factor (TNF) compared with normal donor EC cultures. TNF transiently activates RhoB in ECs coincident with developing leak, and inactivation of RhoB correlates with barrier recovery. The mutation in p190BRhoGAP impairs RhoB inactivation, and the mutant phenotype of patient-derived ECs is replicated by siRNA knockdown of p190BRhoGAP in normal ECs. These data suggest a previously unknown function for p190BRhoGAP in control of capillary EC barrier function that may also be important in acquired systemic capillary leak associated with critical illness in humans.


Subject(s)
Capillary Leak Syndrome/genetics , GTPase-Activating Proteins/genetics , Mutation/genetics , rhoB GTP-Binding Protein/metabolism , Autopsy , Capillary Leak Syndrome/diagnostic imaging , Capillary Leak Syndrome/pathology , Child , Dermis/pathology , Electric Impedance , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Fatal Outcome , Gene Expression Profiling , Humans , Male , Microvessels/pathology , Reproducibility of Results
4.
Nat Med ; 22(9): 1033-1042, 2016 09.
Article in English | MEDLINE | ID: mdl-27548575

ABSTRACT

Cerebral cavernous malformations (CCMs) are vascular malformations that affect the central nervous system and result in cerebral hemorrhage, seizure and stroke. CCMs arise from loss-of-function mutations in one of three genes: KRIT1 (also known as CCM1), CCM2 or PDCD10 (also known as CCM3). PDCD10 mutations in humans often result in a more severe form of the disease relative to mutations in the other two CCM genes, and PDCD10-knockout mice show severe defects, the mechanistic basis for which is unclear. We have recently reported that CCM3 regulates exocytosis mediated by the UNC13 family of exocytic regulatory proteins. Here, in investigating the role of endothelial cell exocytosis in CCM disease progression, we found that CCM3 suppresses UNC13B- and vesicle-associated membrane protein 3 (VAMP3)-dependent exocytosis of angiopoietin 2 (ANGPT2) in brain endothelial cells. CCM3 deficiency in endothelial cells augments the exocytosis and secretion of ANGPT2, which is associated with destabilized endothelial cell junctions, enlarged lumen formation and endothelial cell-pericyte dissociation. UNC13B deficiency, which blunts ANGPT2 secretion from endothelial cells, or treatment with an ANGPT2-neutralizing antibody normalizes the defects in the brain and retina caused by endothelial-cell-specific CCM3 deficiency, including the disruption of endothelial cell junctions, vessel dilation and pericyte dissociation. Thus, enhanced secretion of ANGPT2 in endothelial cells contributes to the progression of CCM disease, providing a new therapeutic approach for treating this devastating pathology.


Subject(s)
Angiopoietin-2/metabolism , Endothelium, Vascular/metabolism , Exocytosis , Hemangioma, Cavernous, Central Nervous System/genetics , Intracellular Signaling Peptides and Proteins/genetics , Angiopoietin-1/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Brain , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression Profiling , Hemangioma, Cavernous, Central Nervous System/metabolism , Humans , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins , Proto-Oncogene Proteins/genetics , Receptor, TIE-2/metabolism , Vesicle-Associated Membrane Protein 3
5.
Circ Res ; 117(2): 121-8, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25940550

ABSTRACT

RATIONALE: The participation of endothelial cells (EC) in many physiological and pathological processes is widely modeled using human EC cultures, but genetic manipulation of these untransformed cells has been technically challenging. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 nuclease (Cas9) technology offers a promising new approach. However, mutagenized cultured cells require cloning to yield homogeneous populations, and the limited replicative lifespan of well-differentiated human EC presents a barrier for doing so. OBJECTIVE: To create a simple but highly efficient method using CRISPR/Cas9 to generate biallelic gene disruption in untransformed human EC. METHODS AND RESULTS: To demonstrate proof-of-principle, we used CRISPR/Cas9 to disrupt the gene for the class II transactivator. We used endothelial colony forming cell-derived EC and lentiviral vectors to deliver CRISPR/Cas9 elements to ablate EC expression of class II major histocompatibility complex molecules and with it, the capacity to activate allogeneic CD4(+) T cells. We show the observed loss-of-function arises from biallelic gene disruption in class II transactivator that leaves other essential properties of the cells intact, including self-assembly into blood vessels in vivo, and that the altered phenotype can be rescued by reintroduction of class II transactivator expression. CONCLUSIONS: CRISPR/Cas9-modified human EC provides a powerful platform for vascular research and for regenerative medicine/tissue engineering.


Subject(s)
CRISPR-Cas Systems , Endothelial Progenitor Cells/cytology , Fetal Blood/cytology , Gene Deletion , Gene Knockout Techniques , Genetic Vectors/pharmacology , Lentivirus/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems/genetics , Cell Separation/methods , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/transplantation , Female , Genes, MHC Class II , Genetic Vectors/drug effects , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/immunology , Humans , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, SCID , Primary Cell Culture/methods , Proteins/genetics , Tetracycline/pharmacology , Vesicular Transport Proteins
6.
PLoS One ; 10(3): e0120075, 2015.
Article in English | MEDLINE | ID: mdl-25816133

ABSTRACT

Capillary leak in severe sepsis involves disruption of endothelial cell tight junctions. We modeled this process by TNF treatment of cultured human dermal microvascular endothelial cell (HDMEC) monolayers, which unlike human umbilical vein endothelial cells form claudin-5-dependent tight junctions and a high-resistance permeability barrier. Continuous monitoring with electrical cell-substrate impedance sensing revealed that TNF disrupts tight junction-dependent HDMEC barriers in discrete steps: an ~5% increase in transendothelial electrical resistance over 40 minutes; a decrease to ~10% below basal levels over 2 hours (phase 1 leak); an interphase plateau of 1 hour; and a major fall in transendothelial electrical resistance to < 70% of basal levels by 8-10 hours (phase 2 leak), with EC50 values of TNF for phase 1 and 2 leak of ~30 and ~150 pg/ml, respectively. TNF leak is reversible and independent of cell death. Leak correlates with disruption of continuous claudin-5 immunofluorescence staining, myosin light chain phosphorylation and loss of claudin-5 co-localization with cortical actin. All these responses require NF-κB signaling, shown by inhibition with Bay 11 or overexpression of IκB super-repressor, and are blocked by H-1152 or Y-27632, selective inhibitors of Rho-associated kinase that do not block other NF-κB-dependent responses. siRNA combined knockdown of Rho-associated kinase-1 and -2 also prevents myosin light chain phosphorylation, loss of claudin-5/actin co-localization, claudin-5 reorganization and reduces phase 1 leak. However, unlike H-1152 and Y-27632, combined Rho-associated kinase-1/2 siRNA knockdown does not reduce the magnitude of phase 2 leak, suggesting that H-1152 and Y-27632 have targets beyond Rho-associated kinases that regulate endothelial barrier function. We conclude that TNF disrupts TJs in HDMECs in two distinct NF-κB-dependent steps, the first involving Rho-associated kinase and the second likely to involve an as yet unidentified but structurally related protein kinase(s).


Subject(s)
Claudin-5/metabolism , Dermis/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , NF-kappa B/metabolism , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Membrane Permeability , Cells, Cultured , Dermis/cytology , Endothelium, Vascular/cytology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Microscopy, Fluorescence , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
7.
Cardiovasc Res ; 100(3): 492-500, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24042014

ABSTRACT

AIM: Angiogenic sprouts arise from microvessels formed by endothelial cells (ECs) invested by pericytes (PCs). The aim of this study was to examine the role of PCs in angiogenic sprouting, an understudied phenomenon. METHODS AND RESULTS: We adapted a human EC spheroid model to examine PC effects on vascular endothelial growth factor-A-induced EC sprouting in vitro by using Bcl-2-transduced human umbilical vein ECs to reduce apoptosis in collagen gels. Human placental PCs, separated from endothelial spheroids by a transwell, or addition of PC-conditioned media increased EC sprouting primarily through hepatocyte growth factor (HGF). Mixed endothelial-PC spheroids formed similar numbers of endothelial sprouts as endothelial spheroids but the sprouts from mixed spheroids were invested by PCs within 24 h. PCs were recruited to the sprouts by platelet-derived growth factor (PDGF)-BB; inhibition of PDGF signalling reduced PC coverage and increased EC sprouting. Transplanted endothelial spheroids give rise to sprouts in vivo that evolve into perfused microvessels. Mixed endothelial-PC spheroids form similar numbers of microvessels as endothelial-only spheroids, but acquire human PC investment and have reduced average lumen diameter. CONCLUSIONS: PCs promote endothelial sprouting by elaborating HGF, but when recruited to invest endothelial sprouts by PDGF-BB, limit the extent of sprouting in vitro and lumen diameter in vivo.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Microvessels/metabolism , Neovascularization, Physiologic , Paracrine Communication , Pericytes/metabolism , Becaplermin , Coculture Techniques , Culture Media, Conditioned/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Microvessels/cytology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Signal Transduction , Spheroids, Cellular , Time Factors , Transfection , Vascular Endothelial Growth Factor A/metabolism
8.
Arterioscler Thromb Vasc Biol ; 33(3): 489-500, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23288152

ABSTRACT

OBJECTIVE: To assess the role claudin-5, an endothelial cell (EC) tight junction protein, plays in establishing basal permeability levels in humans by comparing claudin-5 expression levels in situ and analyzing junctional organization and function in 2 widely used models of cultured ECs, namely human dermal microvascular (HDM)ECs and human umbilical vein (HUV)ECs. METHODS AND RESULTS: By immunofluorescence microscopy, ECs more highly express claudin-5 (but equivalently express vascular endothelial-cadherin) in human dermal capillaries versus postcapillary venules and in umbilical and coronary arteries versus veins, correlating with known segmental differences in tight junction frequencies and permeability barriers. Postconfluent cultured HDMECs express more claudin-5 (but equivalent vascular endothelial-cadherin) and show higher transendothelial electric resistance and lower macromolecular flux than similarly cultured HUVECs. HDMEC junctions are more complex by transmission electron microscopy and show more continuous claudin-5 immunofluorescence than HUVEC junctions. Calcium chelation or dominant negative vascular endothelial-cadherin overexpression decreases transendothelial electric resistance and disrupts junctions in HUVECs, but not in HDMECs. Claudin-5 overexpression in HUVECs fails to increase transendothelial electric resistance or claudin-5 continuity, whereas claudin-5 knockdown in HDMECs, but not in HUVECs, reduces transendothelial electric resistance and increases antibody accessibility to junctional proteins. CONCLUSIONS: Claudin-5 expression and junctional organization control HDMEC and arteriolar-capillary paracellular barriers, whereas HUVEC and venular junctions use vascular endothelial-cadherin.


Subject(s)
Capillary Permeability , Claudin-5/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Skin/blood supply , Tight Junctions/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Calcium/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Claudin-5/genetics , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microvessels/drug effects , Microvessels/ultrastructure , Tight Junctions/drug effects , Tight Junctions/ultrastructure , Time Factors , Transfection
9.
Microcirculation ; 18(2): 102-17, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21166929

ABSTRACT

OBJECTIVE: ECs lining arteries respond to LSS by suppressing pro-inflammatory changes, in part through the activation of MEK5, ERK5 and induction of KLF4. We examined if this anti-inflammatory pathway operates in human ECs lining microvessels, the principal site of inflammatory responses. METHODS: We used immunofluorescence microscopy of human skin to assess ERK5 activation and KLF4 expression in HDMECs in situ. We applied LSS to or overexpressed MEK5/CA in cultured HDMECs and assessed gene expression by microarrays and qRT-PCR and protein expression by Western blotting. We assessed effects of MEK5/CA on TNF responses using qRT-PCR, FACS and measurements of HDMEC monolayer electrical resistance. We used siRNA knockdown to assess the role of ERK5 and KLF4 in these responses. RESULTS: ERK5 phosphorylation and KLF4 expression is observed in HDMECs in situ. LSS activates ERK5 and induces KLF4 in cultured HDMECs. MEK5/CA-transduced HDMECs show activated ERK5 and increased KLF4, thrombomodulin, eNOS, and ICAM-1 expression. MEK5 induction of KLF4 is mediated by ERK5. MEK5/CA-transduced HDMECs are less responsive to TNF, an effect partly mediated by KLF4. CONCLUSIONS: MEK5 activation by LSS inhibits inflammatory responses in microvascular ECs, in part through ERK5-dependent induction of KLF4.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/biosynthesis , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Cells, Cultured , Enzyme Activation , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Shear Strength , Stress, Mechanical
10.
J Invest Dermatol ; 131(1): 17-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21157425

ABSTRACT

During wound healing, dermal macrophages secrete lymphangiogenic vascular endothelial growth factor (VEGF)-C, and lymphatic vessels transport cytokines and cells to draining lymph nodes. In this issue, Moussai et al. show that macrophages in peritumoral nonlesional skin near squamous cell carcinoma secrete prolymphangiogenic VEGF-C. Their study suggests how tumor-associated macrophages and neolymphatic vessels may coordinate metastasis starting early in cutaneous squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/immunology , Lymphangiogenesis/immunology , Macrophages/immunology , Skin Neoplasms/immunology , Vascular Endothelial Growth Factor C/immunology , Carcinoma, Squamous Cell/secondary , Humans , Macrophages/metabolism , Skin/immunology , Skin/metabolism , Skin/pathology , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor C/metabolism
11.
Arterioscler Thromb Vasc Biol ; 30(12): 2553-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20864667

ABSTRACT

OBJECTIVE: The goal of this study was to investigate the novel hypothesis that bone marrow kinase in the X chromosome (Bmx), an established inflammatory mediator of pathological angiogenesis, promotes lymphangiogenesis. METHODS AND RESULTS: We have recently demonstrated a critical role for Bmx in inflammatory angiogenesis. However, the role of Bmx in lymphangiogenesis has not been investigated. Here, we show that in wild-type mice, Bmx is upregulated in lymphatic vessels in response to vascular endothelial growth factor (VEGF). In comparison with wild-type mice, Bmx-deficient mice mount weaker lymphangiogenic responses to VEGF-A and VEGF-C in 2 mouse models. In vitro, Bmx is expressed in cultured human dermal microvascular lymphatic endothelial cells. Furthermore, pharmacological inhibition and short interfering RNA mediated silencing of Bmx reduces VEGF-A and VEGF-C-induced signaling and lymphatic endothelial cell tube formation. Mechanistically, we demonstrated that Bmx differentially regulates VEGFR-2 and VEGFR-3 receptor signaling pathways: Bmx associates with and directly regulates VEGFR-2 activation, whereas Bmx associates with VEGFR-3 and regulates downstream signaling without an effect on the receptor autophosphorylation. CONCLUSIONS: Our in vivo and in vitro results provide the first insight into the mechanism by which Bmx mediates VEGF-dependent lymphangiogenic signaling.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels/enzymology , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Cells, Cultured , Cornea/enzymology , Endothelial Cells/enzymology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , RNA Interference , Recombinant Proteins/metabolism , Signal Transduction , Skin/enzymology , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
12.
J Biol Chem ; 285(31): 23868-79, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20511226

ABSTRACT

TNFR1 (tumor necrosis factor receptor 1) localizes to caveolae of human endothelial-derived EA.hy926 cells. Transduced TNFR1 molecules lacking amino acid residues 229-244 (spanning the transmembrane/intercellular boundary) are expressed on the cell surface equivalently to full-length TNFR1 molecules but incompletely localize to caveolae. A peptide containing this sequence pulls down CAV-1 (caveolin-1) and TNFR1 from cell lysates but fails to do so following disruption of caveolae with methyl-beta-cyclodextrin. We previously reported that methyl-beta-cyclodextrin eliminates caveolae and blocks tumor necrosis factor (TNF)-induced internalization of TNFR1 but not TNF-induced activation of NF-kappaB in EA.hy926 cells. Both CAV-1 and FLOT-2 (flotillin-2), organizing proteins of caveolae and lipid rafts, respectively, associate with caveolae in EA.hy926 cells. Small interfering RNA-mediated knockdown of CAV-1 but not FLOT-2 strikingly reduces caveolae number. Both knockdowns reduce total TNFR1 protein expression, but neither prevents TNFR1 localization to low density membrane domains, TNF-induced internalization of TNFR1, or NF-kappaB activation by TNF. Both CAV-1 and FLOT-2 knockdowns reduce TNF-mediated activation of stress-activated protein kinase (SAPK). However, both knockdowns reduce expression of TRAF2 (TNF receptor-associated factor-2) protein, and small interfering RNA targeting of TRAF2 also selectively inhibits SAPK activation. We conclude that TNFR1 contains a membrane-proximal sequence that targets the receptor to caveolae/lipid rafts. Neither TNFR1 targeting to nor internalization from these low density membrane domains depends upon CAV-1 or FLOT-2. Furthermore, both NF-kappaB and SAPK activation appear independent of both TNFR1 localization to low density membrane domains and to TNF-induced receptor internalization.


Subject(s)
Cell Membrane/metabolism , Endothelial Cells/cytology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Caveolae/metabolism , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Membrane Microdomains/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , NF-kappa B/metabolism , Peptides/chemistry , RNA, Small Interfering/metabolism , Signal Transduction , beta-Cyclodextrins/chemistry
13.
Am J Pathol ; 172(4): 1088-99, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18292233

ABSTRACT

We analyzed tumor necrosis factor (TNF) responses of human umbilical artery and vein endothelial cells (HUAECs and HUVECs) in organ and cell culture. In organ culture, TNF induced expression of E-selectin, VCAM-1, and ICAM-1 on HUVECs but only ICAM-1 on HUAECs. Activation of nuclear factor-kappaB, c-jun, and ATF2 by TNF was comparable in HUAECs and HUVECs, whereas binding of transcription factors and p300 co-activator to the E-selectin enhancer was lower in HUAECs compared to HUVECs. In cell culture, HUAECs rapidly acquired inducible E-selectin and VCAM-1 whereas ICAM-1 inducibility decreased. Culture of HUVECs rapidly decreased TNF responses of all three genes. By 72 hours in cell culture, TNF-treated HUVECs and HUAECs showed comparable adhesion molecule induction and transcription factor binding to the E-selectin enhancer. Freshly isolated HUAECs expressed higher levels of Kruppel-like factor 2 (KLF2) than HUVECs, consistent with greater KLF2 induction by arterial levels of shear stress in vitro. KLF2 expression decreased rapidly in both cell types during culture. Transduction of HUVECs with KLF2 reduced TNF-mediated induction of E-selectin and VCAM-1 while increasing ICAM-1 induction and reduced transcription factor/co-activator binding to the E-selectin enhancer. In conclusion, the differential responses of HUAECs and HUVECs to TNF in organ culture correlate with transcription factor/co-activator binding to DNA and converge during cell culture. Flow-induced expression of KLF2 contributes to the in situ responses of HUAECs but not of HUVECs.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Tumor Necrosis Factors/pharmacology , Umbilical Arteries/drug effects , Umbilical Arteries/metabolism , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , E-Selectin/metabolism , Endothelial Cells/cytology , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , NF-kappa B/metabolism , Organ Culture Techniques , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Transcription Factor AP-1/metabolism , Umbilical Arteries/cytology , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/metabolism
14.
J Immunol ; 180(5): 3467-77, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292573

ABSTRACT

Activation of the classical and noncanonical NF-kappaB pathways by ligation of the lymphotoxin (LT)-beta receptor (LTbetaR) plays a crucial role in lymphoid organogenesis and in the generation of ectopic lymphoid tissue at sites of chronic inflammation. Within these microenvironments, LTbetaR signaling regulates the phenotype of the specialized high endothelial cells. However, the direct effects of LTbetaR ligation on endothelial cells remain unclear. We therefore questioned whether LTbetaR ligation could directly activate endothelial cells and regulate classical and noncanonical NF-kappaB-dependent gene expression. We demonstrate that the LTbetaR ligands LIGHT and LTalpha1beta2 activate both NF-kappaB pathways in HUVECs and human dermal microvascular endothelial cells (HDMEC). Classical pathway activation was less robust than TNF-induced signaling; however, only LIGHT and LTalpha1beta2 and not TNF activated the noncanonical pathway. LIGHT and LTalpha1beta2 induced the expression of classical NF-kappaB-dependent genes in HUVEC, including those encoding the adhesion molecules E-selectin, ICAM-1, and VCAM-1. Consistent with this stimulation, LTbetaR ligation up-regulated T cell adhesion to HUVEC. Furthermore, the homeostatic chemokine CXCL12 was up-regulated by LIGHT and LTalpha1beta2 but not TNF in both HUVEC and HDMEC. Using HUVEC retrovirally transduced with dominant negative IkappaB kinase alpha, we demonstrate that CXCL12 expression is regulated by the noncanonical pathway in endothelial cells. Our findings therefore demonstrate that LTbetaR ligation regulates gene expression in endothelial cells via both NF-kappaB pathways and we identify CXCL12 as a bona fide noncanonical NF-kappaB-regulated gene in these cells.


Subject(s)
Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Gene Expression Regulation/immunology , Inflammation Mediators/physiology , Lymphotoxin alpha1, beta2 Heterotrimer/physiology , NF-kappa B/physiology , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Line , Endothelium, Vascular/metabolism , Humans , Ligands , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Lymphotoxin beta Receptor/biosynthesis , Lymphotoxin beta Receptor/metabolism , Lymphotoxin beta Receptor/physiology , Lymphotoxin-alpha/metabolism , Lymphotoxin-alpha/physiology , Lymphotoxin-beta/metabolism , Lymphotoxin-beta/physiology , NF-kappa B/metabolism , Signal Transduction/genetics , Skin/blood supply , Skin/cytology , Skin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Up-Regulation/genetics , Up-Regulation/immunology
15.
Nucleic Acids Res ; 36(4): 1081-97, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18096615

ABSTRACT

Tumor necrosis factor (TNF) initiates local inflammation by triggering endothelial cells (EC) to express adhesion molecules for leukocytes such as intercellular adhesion molecule-1 (ICAM-1 or CD54). A prior study identified siRNA molecules that reduce ICAM-1 expression in cultured human umbilical vein EC (HUVEC). One of these, ISIS 121736, unexpectedly inhibits TNF-mediated up-regulation of additional molecules on EC, including E-selectin (CD62E), VCAM-1 (CD106) and HLA-A,B,C. 736 siRNA transfection was not toxic for EC nor was there any evidence of an interferon response. 736 Transfection of EC blocked multiple early TNF-related signaling events, including activation of NF-kappaB. IL-1 activation of these same pathways was not inhibited. A unifying explanation is that 736 siRNA specifically reduced expression of mRNA encoding tumor necrosis factor receptor 1 (TNFR1) as well as TNFR1 surface expression. A sequence with high identity to the 736 antisense strand (17 of 19 bases) is present within the 3'UTR of human TNFR1 mRNA. An EGFP construct incorporating the 3'UTR of TNFR1 was silenced by 736 siRNA and this effect was lost by mutagenesis of this complementary sequence. Chemical modification and mismatches within the sense strand of 736 also inhibited silencing activity. In summary, an siRNA molecule selected to target ICAM-1 through its antisense strand exhibited broad anti-TNF activities. We show that this off-target effect is mediated by siRNA knockdown of TNFR1 via its sense strand. This may be the first example in which the off-target effect of an siRNA is actually responsible for the anticipated effect by acting to reduce expression of a protein (TNFR1) that normally regulates expression of the intended target (ICAM-1).


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , RNA Interference , RNA, Small Interfering/chemistry , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , 3' Untranslated Regions/chemistry , Cells, Cultured , Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Interleukin-1/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sequence Alignment , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
16.
J Invest Dermatol ; 127(4): 762-74, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17195014

ABSTRACT

Tumor necrosis factor (TNF)-induced ICAM-1 in endothelial cells (EC) promotes leukocyte adhesion. Here we report that ICAM-1 also effects EC barrier function. Control- or E-selectin-transduced human dermal microvascular EC (HDMEC) form a barrier to flux of proteins and to passage of current (measured as transendothelial electrical resistance or TEER). HDMEC transduced with ICAM-1 at levels comparable to that induced by TNF show reduced TEER, but do so without overtly changing their cell junctions, cell shape, or cytoskeleton organization. Higher levels of ICAM-1 further reduce TEER, increase F/G-actin ratios, rearrange the actin cytoskeleton to cause cell elongation, and alter junctional zona occludens 1 and vascular endothelial-cadherin staining. Transducing with ICAM-1 lacking an intracellular region also reduces TEER. TNF-induced changes in TEER and shape follow a similar time course as ICAM-1 induction; however, the fall in TEER occurs at lower TNF concentrations. Inhibiting NF-kappaB activation blocks ICAM-1 induction; TEER reduction, and shape change. Specific small-interfering RNA knockdown of ICAM-1 partially inhibits TNF-induced shape change. We conclude that moderately elevated ICAM-1 expression reduces EC barrier function and that expressing higher levels of ICAM-1 affects cell junctions and the cytoskeleton. Induction of ICAM-1 may contribute to but does not fully account for TNF-induced vascular leak and EC shape change.


Subject(s)
Capillary Permeability/physiology , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Skin/blood supply , Adherens Junctions/ultrastructure , Capillary Permeability/drug effects , Cell Shape/drug effects , Cells, Cultured , Cytoskeleton/ultrastructure , Electric Impedance , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Junctions/physiology , Microcirculation , Tight Junctions/ultrastructure , Transduction, Genetic , Tumor Necrosis Factor-alpha/pharmacology
17.
Eur J Immunol ; 37(1): 177-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17163451

ABSTRACT

Effects of IL-10 on endothelium-dependent T cell activation have not been investigated in detail. We confirm expression of the IL-10 receptor and effective signaling via STAT-3 in human umbilical vein endothelial cells (HUVEC). In CD4 T cell cocultures with HUVEC, pretreatment of endothelial cells with IL-10 resulted in significant dose-dependent inhibition of CD4 T cell proliferation, which also occurred when IL-10 was removed after pretreatment before starting cocultures. Th1/Th2 polarization of proliferated T cells, endothelial nitric oxide (NO), or IL-12 production were unchanged. However, IL-10 stimulation resulted in up-regulation of SOCS-3, a negative regulator of cytokine secretion, and induction of the inhibitory surface molecules immunoglobulin-like transcript 3 and 4 (ILT3/ILT4) in EC, potentially involving glucocorticoid-induced leucine zipper (GILZ). Addition of blocking antibodies against ILT3/ILT4 to EC/T cell cocultures resulted in nearly complete reestablishment of T cell proliferation. In contrast, addition of soluble ILT3 or overexpression of ILT3 in cocultures significantly reduced T cell proliferation. No induction of foxp3+ regulatory T cells was seen. In conclusion, the T cell costimulatory potential of human EC is markedly suppressed by IL-10 due to up-regulation of ILT3/ILT4, obviously not involving generation of Treg. This identifies a novel action of IL-10 in EC and a potential therapeutical target for local immunomodulation.


Subject(s)
Endothelium, Vascular/immunology , Growth Inhibitors/physiology , Interleukin-10/physiology , Lymphocyte Activation/immunology , Membrane Glycoproteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Immunologic/biosynthesis , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Receptors, Immunologic/physiology , Receptors, Interleukin-10/biosynthesis , T-Lymphocyte Subsets/metabolism
18.
Circulation ; 114(12): 1293-300, 2006 Sep 19.
Article in English | MEDLINE | ID: mdl-16940188

ABSTRACT

BACKGROUND: Heparin, used clinically as an anticoagulant, also has antiinflammatory properties and has been described to inhibit interferon (IFN)-gamma responses in endothelial cells. We investigated the effects of heparin on the IFN-gamma-inducible chemokines IP-10/CXCL10, I-TAC/CXCL11, and Mig/CXCL9, which play important roles in the vascular recruitment of IFN-gamma-producing Th1 cells through interactions with their cognate receptor, CXCR3. METHODS AND RESULTS: Patients undergoing coronary artery bypass grafting were studied because coronary atherosclerosis is recognized as a Th1-type inflammatory disease and the subjects required systemic heparinization. Plasma levels of IP-10, I-TAC, and Mig increased immediately after heparin administration and diminished promptly after heparin antagonism with protamine. These effects were independent of detectable circulating IFN-gamma or the IFN-gamma inducer interleukin-12. We confirmed previous reports that heparin inhibits the IFN-gamma-dependent production of CXCR3 chemokine ligands using atherosclerotic coronary arteries in organ culture. In addition to prolonged treatment decreasing chemokine secretion, heparin rapidly displaced membrane-associated IP-10 from cultured endothelial cells that did not express CXCR3 and reduced the IP-10-dependent transendothelial migration of T helper cells under conditions of venular shear stress. Finally, heparin administration to immunodeficient mouse hosts decreased both the recruitment and accumulation of memory T cells within allogeneic human coronary arteries. CONCLUSIONS: Besides inhibiting IFN-gamma responses, heparin has further immunomodulatory effects by competing for binding with IP-10, I-TAC, and Mig on endothelial cells. Disruption of CXCR3+ Th1 cell trafficking to arteriosclerotic arteries may contribute to the therapeutic efficacy of heparin in inflammatory arterial diseases, and nonanticoagulant heparin derivatives may represent a novel antiinflammatory strategy.


Subject(s)
Chemokines/metabolism , Coronary Vessels/pathology , Endothelium, Vascular/metabolism , Heparin/metabolism , Heparin/pharmacology , Interferon-gamma/metabolism , T-Lymphocytes/pathology , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Heparin Antagonists/pharmacology , Humans , Interleukin-12/metabolism , Protamines/pharmacology , Protein Binding , Receptors, CXCR3 , Receptors, Chemokine/drug effects , Receptors, Chemokine/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Th1 Cells/pathology
19.
Transplantation ; 82(1 Suppl): S9-14, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16829798

ABSTRACT

The chemokine interferon (IFN)-gamma-inducible protein of 10 kDa (IP-10; CXCL10) has been implicated in recruitment of T cells into rejecting allografts yet appears ineffective at stimulating human peripheral blood CD4 T cells to transmigrate across tumor necrosis factor (TNF)-treated human endothelial cell (EC) monolayers in vitro. The same cells rapidly (within 15 min) transmigrate across TNF-treated EC monolayers overlaid with stromal cell-derived factor-1 alpha (SDF-1 alpha) and subjected to shear stress. The effector memory subset within the CD4 T cell population, defined as CD45RO, CD62L and CCR7, which constitutes less than 10% of total CD4 T cells, does respond to IP-10 but requires enrichment to be observed in this model. Central memory T cells do not respond to IP-10. Transendothelial migration of effector memory CD4 T cells requires TNF-pretreatment of the EC monolayer and application of venular shear force during the assay. TNF treatment of ECs may be effectively replaced by transduction of vascular cell adhesion molecule-1 or intercellular adhesion molecule-1 but not E-selectin.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Communication/immunology , Chemokines, CXC/pharmacology , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Graft Rejection/immunology , T-Lymphocyte Subsets/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Movement/genetics , Cells, Cultured , Chemokine CXCL10 , Chemokine CXCL12 , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Graft Rejection/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/physiology , L-Selectin/analysis , Leukocyte Common Antigens/analysis , Receptors, CCR7 , Receptors, Chemokine/analysis , Stress, Mechanical , T-Lymphocyte Subsets/immunology , Transduction, Genetic , Tumor Necrosis Factors/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/physiology
20.
Adv Dermatol ; 20: 163-201, 2004.
Article in English | MEDLINE | ID: mdl-15544200

ABSTRACT

During inflammation, coordinated expression of cytokine-induced adhesion molecules (CAMs) on postcapillary venular endothelial cells (ECs) regulates leukocyte recruitment. During their recruitment from blood, leukocytes adhere to EC CAMs, activating signaling pathways inside ECs. In a forthcoming paradigm, leukocyte transendothelial migration requires active EC participation, with extracellular adhesive CAM functions mirrored by cytoplasmic do-main-dependent intracellular events. These events serve to reorganize the EC actin cytoskeleton. Investigators have visualized this as changes in EC shape, transient opening of EC-EC contacts, and redistribution of CAMs expressed on the luminal EC surface. In this review, we (1) summarize the overlapping extracellular adhesive properties of the 3 EC CAMs most important for leukocyte recruitment during inflammation, namely, E-selectin, vascular cell adhesion molecule, and intercellular adhesion molecule-1; (2) explore the role of these 3 CAMs as signal transducers by identifying the intracellular signals (Ca++, Rho/Rac, and phosphatidylinositol 4,5-bisphosphate) that upon leukocyte engagement, reorganize the EC cytoskeleton and redistribute these apical CAMs, thereby favoring leukocyte recruitment; and (3) describe how CAM-derived signals lead to ezrin-radixin-moesin complex formation and how this complex of plasma membrane-cytoskeleton adapter proteins coordinates CAM-driven intracellular signals with extracellular adhesive CAM functions. This literature review suggests that the cytoplasmic domains of these EC CAMs and their downstream effectors represent new and potentially beneficial intracellular therapeutic targets for treating diseases of the skin.


Subject(s)
Endothelium, Vascular/physiology , Inflammation/physiopathology , Leukocytes/physiology , Skin Diseases/physiopathology , Cell Adhesion/physiology , Humans , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL