Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell ; 186(11): 2392-2409.e21, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37164012

ABSTRACT

T cell responses play an important role in protection against beta-coronavirus infections, including SARS-CoV-2, where they associate with decreased COVID-19 disease severity and duration. To enhance T cell immunity across epitopes infrequently altered in SARS-CoV-2 variants, we designed BNT162b4, an mRNA vaccine component that is intended to be combined with BNT162b2, the spike-protein-encoding vaccine. BNT162b4 encodes variant-conserved, immunogenic segments of the SARS-CoV-2 nucleocapsid, membrane, and ORF1ab proteins, targeting diverse HLA alleles. BNT162b4 elicits polyfunctional CD4+ and CD8+ T cell responses to diverse epitopes in animal models, alone or when co-administered with BNT162b2 while preserving spike-specific immunity. Importantly, we demonstrate that BNT162b4 protects hamsters from severe disease and reduces viral titers following challenge with viral variants. These data suggest that a combination of BNT162b2 and BNT162b4 could reduce COVID-19 disease severity and duration caused by circulating or future variants. BNT162b4 is currently being clinically evaluated in combination with the BA.4/BA.5 Omicron-updated bivalent BNT162b2 (NCT05541861).


Subject(s)
BNT162 Vaccine , COVID-19 , Animals , Cricetinae , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Epitopes , SARS-CoV-2/genetics
2.
Cancer Cell ; 40(9): 1010-1026.e11, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36027916

ABSTRACT

Neoantigens arising from mutations in tumor DNA provide targets for immune-based therapy. Here, we report the clinical and immune data from a Phase Ib clinical trial of a personalized neoantigen-vaccine NEO-PV-01 in combination with pemetrexed, carboplatin, and pembrolizumab as first-line therapy for advanced non-squamous non-small cell lung cancer (NSCLC). This analysis of 38 patients treated with the regimen demonstrated no treatment-related serious adverse events. Multiple parameters including baseline tumor immune infiltration and on-treatment circulating tumor DNA levels were highly correlated with clinical response. De novo neoantigen-specific CD4+ and CD8+ T cell responses were observed post-vaccination. Epitope spread to non-vaccinating neoantigens, including responses to KRAS G12C and G12V mutations, were detected post-vaccination. Neoantigen-specific CD4+ T cells generated post-vaccination revealed effector and cytotoxic phenotypes with increased CD4+ T cell infiltration in the post-vaccine tumor biopsy. Collectively, these data support the safety and immunogenicity of this regimen in advanced non-squamous NSCLC.


Subject(s)
Cancer Vaccines , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antigens, Neoplasm , CD8-Positive T-Lymphocytes , Cancer Vaccines/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics
3.
Cell ; 183(2): 347-362.e24, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33064988

ABSTRACT

Neoantigens arise from mutations in cancer cells and are important targets of T cell-mediated anti-tumor immunity. Here, we report the first open-label, phase Ib clinical trial of a personalized neoantigen-based vaccine, NEO-PV-01, in combination with PD-1 blockade in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. This analysis of 82 patients demonstrated that the regimen was safe, with no treatment-related serious adverse events observed. De novo neoantigen-specific CD4+ and CD8+ T cell responses were observed post-vaccination in all of the patients. The vaccine-induced T cells had a cytotoxic phenotype and were capable of trafficking to the tumor and mediating cell killing. In addition, epitope spread to neoantigens not included in the vaccine was detected post-vaccination. These data support the safety and immunogenicity of this regimen in patients with advanced solid tumors (Clinicaltrials.gov: NCT02897765).


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Precision Medicine/methods , Aged , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/drug therapy , Melanoma/immunology , Middle Aged , Mutation , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology
4.
Blood ; 130(23): 2548-2558, 2017 12 07.
Article in English | MEDLINE | ID: mdl-28899852

ABSTRACT

Hemostasis in vertebrates involves both a cellular and a protein component. Previous studies in jawless vertebrates (cyclostomes) suggest that the protein response, which involves thrombin-catalyzed conversion of a soluble plasma protein, fibrinogen, into a polymeric fibrin clot, is conserved in all vertebrates. However, similar data are lacking for the cellular response, which in gnathostomes is regulated by von Willebrand factor (VWF), a glycoprotein that mediates the adhesion of platelets to the subendothelial matrix of injured blood vessels. To gain evolutionary insights into the cellular phase of coagulation, we asked whether a functional vwf gene is present in the Atlantic hagfish, Myxine glutinosa We found a single vwf transcript that encodes a simpler protein compared with higher vertebrates, the most striking difference being the absence of an A3 domain, which otherwise binds collagen under high-flow conditions. Immunohistochemical analyses of hagfish tissues and blood revealed Vwf expression in endothelial cells and thrombocytes. Electron microscopic studies of hagfish tissues demonstrated the presence of Weibel-Palade bodies in the endothelium. Hagfish Vwf formed high-molecular-weight multimers in hagfish plasma and in stably transfected CHO cells. In functional assays, botrocetin promoted VWF-dependent thrombocyte aggregation. A search for vwf sequences in the genome of sea squirts, the closest invertebrate relatives of hagfish, failed to reveal evidence of an intact vwf gene. Together, our findings suggest that VWF evolved in the ancestral vertebrate following the divergence of the urochordates some 500 million years ago and that it acquired increasing complexity though sequential insertion of functional modules.


Subject(s)
Hagfishes , von Willebrand Factor/genetics , von Willebrand Factor/metabolism , ADAMTS13 Protein/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cloning, Molecular , Cricetulus , DNA, Complementary , Endothelium, Vascular/metabolism , Evolution, Molecular , Gene Expression , Homeostasis , Humans , Models, Molecular , Platelet Aggregation , Protein Conformation , Protein Domains , Protein Folding , Protein Multimerization , Protein Processing, Post-Translational , Protein Transport , Proteolysis , Structure-Activity Relationship , Vertebrates , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/ultrastructure , von Willebrand Factor/chemistry
5.
Am J Pathol ; 187(10): 2337-2347, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28736316

ABSTRACT

Tumors induce their heterogeneous vasculature by secreting vascular endothelial growth factor (VEGF)-A. Anti-VEGF/VEGF receptor (VEGFR) drugs treat cancer, but the underlying mechanisms remain unclear. An adenovirus expressing VEGF-A (Ad-VEGF-A164) replicates the tumor vasculature in mice without tumor cells. Mother vessels (MV) are the first angiogenic vessel type to form in tumors and after Ad-VEGF-A164. Multiday treatments with a VEGF trap reverted MV back to normal microvessels. We now show that, within hours, a single dose of several anti-VEGF drugs collapsed MV to form glomeruloid microvascular proliferations (GMP), accompanied by only modest endothelial cell death. GMP, common in many human cancers but of uncertain origin, served as an intermediary step in MV reversion to normal microvessels. The vasodisruptive drug combretastatin CA4 also targeted MV selectively but acted differently, extensively killing MV endothelium. Antivascular changes were quantified with a novel Evans blue dye assay that measured vascular volumes. As in tumors, Ad-VEGF-A164 strikingly increased endothelial nitric oxide synthase (eNOS) expression. The eNOS inhibitor N(G)-Nitro-l-arginine methyl ester mimicked anti-VEGF/VEGFR drugs, rapidly collapsing MV to GMP. Inhibition of eNOS reduces synthesis of its vasodilatory product, nitric oxide, leading to arterial contraction. Patients and mice receiving anti-VEGF/VEGFR drugs develop hypertension, reflecting systemic arterial contraction. Together, anti-VEGF/VEGFR drugs act in part by inhibiting eNOS, causing vasocontraction, MV collapse to GMP, and subsequent reversion of GMP to normal microvessels, all without extensive vascular killing.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Blood Vessels/drug effects , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenoviridae/metabolism , Animals , Bibenzyls/pharmacology , Cell Death/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Hypertension/pathology , Mice, Inbred C57BL , Mice, Nude , Microvessels/drug effects , Microvessels/pathology , Models, Biological , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Biochem Biophys Res Commun ; 465(3): 338-43, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26241677

ABSTRACT

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane-associated glycoprotein that is highly and selectively expressed on the plasma membranes of tumor cells, cultured endothelial cells, and, in vivo, on tumor-associated endothelium. Immunofluorescence microscopy also demonstrated TM4SF1 in cytoplasm and, tentatively, within nuclei. With monoclonal antibody 8G4, and the finer resolution afforded by immuno-nanogold transmission electron microscopy, we now demonstrate TM4SF1 in uncoated cytoplasmic vesicles, nuclear pores and nucleoplasm. Because of its prominent surface location on tumor cells and tumor-associated endothelium, TM4SF1 has potential as a dual therapeutic target using an antibody drug conjugate (ADC) approach. For ADC to be successful, antibodies reacting with cell surface antigens must be internalized for delivery of associated toxins to intracellular targets. We now report that 8G4 is efficiently taken up into cultured endothelial cells by uncoated vesicles in a dynamin-dependent, clathrin-independent manner. It is then transported along microtubules through the cytoplasm and passes through nuclear pores into the nucleus. These findings validate TM4SF1 as an attractive candidate for cancer therapy with antibody-bound toxins that have the capacity to react with either cytoplasmic or nuclear targets in tumor cells or tumor-associated vascular endothelium.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Clathrin/immunology , Dynamins/immunology , Endothelial Cells/immunology , Neoplasm Proteins/immunology , Subcellular Fractions/immunology , Cells, Cultured , Endothelial Cells/ultrastructure , Humans
7.
Angiogenesis ; 17(4): 897-907, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24986520

ABSTRACT

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane glycoprotein that regulates cell motility and proliferation. TM4SF1 is an attractive cancer target because of its high expression in both tumor cells and on the vascular endothelial cells lining tumor blood vessels. We generated mouse monoclonal antibodies against human TM4SF1 in order to evaluate their therapeutic potential; 13 of the antibodies we generated reacted with extracellular loop-2 (EL2), TM4SF1's larger extracellular, lumen-facing domain. However, none of these antibodies reacted with mouse TM4SF1, likely because the EL2 of mouse TM4SF1 differs significantly from that of its human counterpart. Therefore, to test our antibodies in vivo, we employed an established model of engineered human vessels in which human endothelial colony-forming cells (ECFC) and human mesenchymal stem cells (MSC) are incorporated into Matrigel plugs that are implanted subcutaneously in immunodeficient nude mice. We modified the original protocol by (1) preculturing human ECFC on laminin, fibronectin, and collagen-coated plates, and (2) increasing the ECFC/MSC ratio. These modifications significantly increased the human vascular network in Matrigel implants. Two injections of one of our anti-TM4SF1 EL2 monoclonal antibodies, 8G4, effectively eliminated the human vascular component present in these plugs; they also abrogated human PC3 prostate cancer cells that were incorporated into the ECFC/MSC Matrigel mix. Together, these studies provide a mouse model for assessing tumor xenografts that are supplied by a human vascular network and demonstrate that anti-TM4SF1 antibodies such as 8G4 hold promise for cancer therapy.


Subject(s)
Antigens, Surface/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Neovascularization, Pathologic , Tissue Engineering/methods , Transcription, Genetic
8.
J Cell Sci ; 127(Pt 17): 3720-34, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25002404

ABSTRACT

Immune cell trafficking requires the frequent breaching of the endothelial barrier either directly through individual cells ('transcellular' route) or through the inter-endothelial junctions ('paracellular' route). What determines the loci or route of breaching events is an open question with important implications for overall barrier regulation. We hypothesized that basic biomechanical properties of the endothelium might serve as crucial determinants of this process. By altering junctional integrity, cytoskeletal morphology and, consequently, local endothelial cell stiffness of different vascular beds, we could modify the preferred route of diapedesis. In particular, high barrier function was associated with predominantly transcellular migration, whereas negative modulation of junctional integrity resulted in a switch to paracellular diapedesis. Furthermore, we showed that lymphocytes dynamically probe the underlying endothelium by extending invadosome-like protrusions (ILPs) into its surface that deform the nuclear lamina, distort actin filaments and ultimately breach the barrier. Fluorescence imaging and pharmacologic depletion of F-actin demonstrated that lymphocyte barrier breaching efficiency was inversely correlated with local endothelial F-actin density and stiffness. Taken together, these data support the hypothesis that lymphocytes are guided by the mechanical 'path of least resistance' as they transverse the endothelium, a process we term 'tenertaxis'.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Endothelial Cells/metabolism , Lymphocytes/metabolism , Animals , Biomechanical Phenomena , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Leukocytes/metabolism , Lymphocytes/cytology , Rats
9.
Stem Cells ; 30(11): 2472-86, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22887987

ABSTRACT

Systemically administered adult mesenchymal stem cells (MSCs), which are being explored in clinical trials to treat inflammatory disease, exhibit the critical ability to extravasate at sites of inflammation. We aimed to characterize the basic cellular processes mediating this extravasation and compare them to those involved in leukocyte transmigration. Using high-resolution confocal and dynamic microscopy, we show that, like leukocytes, human bone marrow-derived MSC preferentially adhere to and migrate across tumor necrosis factor-α-activated endothelium in a vascular cell adhesion molecule-1 (VCAM-1) and G-protein-coupled receptor signaling-dependent manner. As several studies have suggested, we observed that a fraction of MSC was integrated into endothelium. In addition, we observed two modes of transmigration not previously observed for MSC: Paracellular (between endothelial cells) and transcellular (directly through individual endothelial cells) diapedesis through discrete gaps and pores in the endothelial monolayer, in association with VCAM-1-enriched "transmigratory cups". Contrasting leukocytes, MSC transmigration was not preceded by significant lateral migration and occurred on the time scale of hours rather than minutes. Interestingly, rather than lamellipodia and invadosomes, MSC exhibited nonapoptotic membrane blebbing activity that was similar to activities previously described for metastatic tumor and embryonic germ cells. Our studies suggest that low avidity binding between endothelium and MSC may grant a permissive environment for MSC blebbing. MSC blebbing was associated with early stages of transmigration, in which blebs could exert forces on underlying endothelial cells indicating potential functioning in breaching the endothelium. Collectively, our data suggest that MSC transmigrate actively into inflamed tissues via both leukocyte-like and novel mechanisms.


Subject(s)
Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Transendothelial and Transepithelial Migration , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Adhesion , Cell Membrane/metabolism , Cell-Derived Microparticles/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Leukocytes/physiology , Mesenchymal Stem Cells/ultrastructure , Microvessels/cytology , Rats , Vascular Cell Adhesion Molecule-1/metabolism
10.
Blood ; 120(11): 2167-73, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22674805

ABSTRACT

VEGF induces vascular permeability (VP) in ischemic diseases and cancer, leading to many pathophysiological consequences. The molecular mechanisms by which VEGF acts to induce hyperpermeability are poorly understood and in vivo models that easily facilitate real-time, genetic studies of VP do not exist. In the present study, we report a heat-inducible VEGF transgenic zebrafish (Danio rerio) model through which VP can be monitored in real time. Using this approach with morpholino-mediated gene knock-down and knockout mice, we describe a novel role of phospholipase Cß3 as a negative regulator of VEGF-mediated VP by regulating intracellular Ca2+ release. Our results suggest an important effect of PLCß3 on VP and provide a new model with which to identify genetic regulators of VP crucial to several disease processes.


Subject(s)
Capillary Permeability , Endothelium, Vascular/metabolism , Phospholipase C beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Genetically Modified , Calcium Signaling/drug effects , Capillary Permeability/drug effects , Cells, Cultured , Embryo, Nonmammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response , High-Throughput Screening Assays , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Morpholinos/pharmacology , Phospholipase C beta/antagonists & inhibitors , Phospholipase C beta/genetics , Promoter Regions, Genetic/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
J Immunol ; 188(8): 3686-99, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22442443

ABSTRACT

Adaptive immunity requires that T cells efficiently scan diverse cell surfaces to identify cognate Ag. However, the basic cellular mechanisms remain unclear. In this study, we investigated this process using vascular endothelial cells, APCs that possess a unique and extremely advantageous, planar morphology. High-resolution imaging revealed that CD4 memory/effector T cells dynamically probe the endothelium by extending submicron-scale, actin-rich "invadosome/podosome-like protrusions" (ILPs). The intimate intercellular contacts enforced by ILPs consistently preceded and supported T cell activation in response to endothelial MHC class II/Ag. The resulting calcium flux stabilized dense arrays of ILPs (each enriched in TCR, protein kinase C-θ, ZAP70, phosphotyrosine, and HS1), forming what we term a podo-synapse. Similar findings were made using CD8 CTLs on endothelium. Furthermore, careful re-examination of both traditional APC models and professional APCs suggests broad relevance for ILPs in facilitating Ag recognition. Together, our results indicate that ILPs function as sensory organelles that serve as actuators of immune surveillance.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Surface Extensions/immunology , Endothelial Cells/immunology , Adaptive Immunity , Animals , Antigen Presentation , Antigen-Presenting Cells/cytology , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CHO Cells , Calcium/immunology , Calcium/metabolism , Calcium Signaling , Cell Communication/immunology , Cell Surface Extensions/ultrastructure , Cricetinae , Endothelial Cells/cytology , Genes, MHC Class II , Humans , Immunologic Memory , Lymphocyte Activation , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Transendothelial and Transepithelial Migration , Transfection
12.
Angiogenesis ; 14(3): 345-54, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21626280

ABSTRACT

Transmembrane-4-L-six-family-1 (TM4SF1) is a tetraspanin-like membrane protein that is highly and selectively expressed by cultured endothelial cells (EC) and, in vivo, by EC lining angiogenic tumor blood vessels. TM4SF1 is necessary for the formation of unusually long (up to a 50 µm), thin (~100-300 nm wide), F-actin-poor EC cell projections that we term 'nanopodia'. Immunostaining of nanopodia at both the light and electron microsopic levels localized TM4SF1 in a regularly spaced, banded pattern, forming TM4FS1-enriched domains. Live cell imaging of GFP-transduced HUVEC demonstrated that EC project nanopodia as they migrate and interact with neighboring cells. When TM4SF1 mRNA levels in EC were increased from the normal ~90 mRNA copies/cell to ~400 copies/cell through adenoviral transduction, EC projected more and longer nanopodia from the entire cell circumference but were unable to polarize or migrate effectively. When fibroblasts, which normally express TM4SF1 at ~5 copies/cell, were transduced to express TM4SF1 at EC-like levels, they formed typical TM4SF1-banded nanopodia, and broadened, EC-like lamellipodia. Mass-spectrometry demonstrated that TM4SF1 interacted with myosin-10 and ß-actin, proteins involved in filopodia formation and cell migration. In summary, TM4SF1, like genuine tetraspanins, serves as a molecular organizer that interacts with membrane and cytoskeleton-associated proteins and uniquely initiates the formation of nanopodia and facilitates cell polarization and migration.


Subject(s)
Antigens, Surface/metabolism , Cell Membrane Structures/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Endothelial Cells/metabolism , Fibroblasts/metabolism , Neoplasm Proteins/metabolism , Pseudopodia/metabolism , Antigens, Surface/genetics , Cell Membrane Structures/genetics , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/metabolism , Endothelial Cells/cytology , Fibroblasts/cytology , Humans , Neoplasm Proteins/genetics , Pseudopodia/genetics
13.
Am J Pathol ; 175(4): 1768-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19729487

ABSTRACT

Caveolin-1, the signature protein of endothelial cell caveolae, has many important functions in vascular cells. Caveolae are thought to be the transcellular pathway by which plasma proteins cross normal capillary endothelium, but, unexpectedly, cav-1(-/-) mice, which lack caveolae, have increased permeability to plasma albumin. The acute increase in vascular permeability induced by agents such as vascular endothelial growth factor (VEGF)-A occurs through venules, not capillaries, and particularly through the vesiculo-vacuolar organelle (VVO), a unique structure composed of numerous interconnecting vesicles and vacuoles that together span the venular endothelium from lumen to ablumen. Furthermore, the hyperpermeable blood vessels found in pathological angiogenesis, mother vessels, are derived from venules. The present experiments made use of cav-1(-/-) mice to investigate the relationship between caveolae and VVOs and the roles of caveolin-1 in VVO structure in the acute vascular hyperpermeability induced by VEGF-A and in pathological angiogenesis and associated chronic vascular hyperpermeability. We found that VVOs expressed caveolin-1 variably but, in contrast to caveolae, were present in normal numbers and with apparently unaltered structure in cav-1(-/-) mice. Nonetheless, VEGF-A-induced hyperpermeability was strikingly reduced in cav-1(-/-) mice, as was pathological angiogenesis and associated chronic vascular hyperpermeability, whether induced by VEGF-A(164) or by a tumor. Thus, caveolin-1 is not necessary for VVO structure but may have important roles in regulating VVO function in acute vascular hyperpermeability and angiogenesis.


Subject(s)
Capillary Permeability/physiology , Caveolin 1/deficiency , Neovascularization, Pathologic/physiopathology , Adenoviridae , Animals , Caveolin 1/metabolism , Cell Line, Tumor , Cell Proliferation , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Female , Mice , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Skin/blood supply , Skin/pathology , Skin/ultrastructure , Vacuoles/metabolism , Vacuoles/ultrastructure , Vascular Endothelial Growth Factor A/metabolism
14.
Immunity ; 26(6): 784-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17570692

ABSTRACT

Diapedesis is critical for immune system function and inflammatory responses. This occurs by migration of blood leukocytes either directly through individual microvascular endothelial cells (the "transcellular" route) or between them (the "paracellular" route). Mechanisms for transcellular pore formation in endothelium remain unknown. Here we demonstrate that lymphocytes used podosomes and extended "invasive podosomes" to palpate the surface of, and ultimately form transcellular pores through, the endothelium. In lymphocytes, these structures were dependent on Src kinase and the actin regulatory protein WASP; inhibition of podosome formation selectively blocked the transcellular route of diapedesis. In endothelium, membrane fusion events dependent on the SNARE-containing membrane fusion complex and intracellular calcium were required for efficient transcellular pore formation in response to podosomes. These findings provide insights into basic mechanisms for leukocyte trafficking and the functions of podosomes.


Subject(s)
Cell Movement , Endothelium, Vascular/immunology , Lymphocytes/immunology , Pseudopodia/physiology , Capillaries/cytology , Capillaries/immunology , Cell Membrane/chemistry , Endothelium, Vascular/cytology , Humans , Leukocytes/immunology , Lymphocytes/ultrastructure , Membrane Fusion , Porosity , Pseudopodia/ultrastructure , SNARE Proteins/analysis , SNARE Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , src-Family Kinases/metabolism
15.
Immunol Lett ; 99(1): 122-9, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15894120

ABSTRACT

Mast cells are important effector cells in IgE-associated immune responses, but also can contribute to host defense in certain examples of bacterial infection. We found that genetically mast cell-deficient WBB6F1-Kit(W)/Kit(W-v) mice exhibited more bacterial CFUs per spleen by 6 days after intraperitoneal injection of bioluminescent Salmonella typhimurium, and died more rapidly after infection, than did the congenic WBB6F1-Kit(+/+) wild type mice. Adoptive transfer of bone marrow-derived cultured mast cells of Kit(+/+) origin to the peritoneal cavity of Kit(W)/Kit(W-v) mice resulted in engraftment of mast cells in the peritoneal cavity and mesentery of the recipient mice, and the development of large numbers of mast cells in the spleen. However, such mast cell-engrafted Kit(W)/Kit(W-v) mice appeared sicker after intraperitoneal injection with S. typhimurium than did mast cell-deficient Kit(W)/Kit(W-v) mice, and exhibited numbers of CFUs of bacteria per spleen, and a survival curve, that were not significantly different than those of Kit(W)/Kit(W-v) mice. These results, when taken together with prior studies investigating the roles of mast cells in innate immunity, strongly suggest that whether mast cells can be shown to have a significant role in enhancing survival during bacterial infections may depend critically on the details of the particular experimental systems examined.


Subject(s)
Mast Cells/transplantation , Proto-Oncogene Proteins c-kit/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Animals , Cell Count , Disease Models, Animal , Female , Injections, Intraperitoneal , Luminescent Measurements , Mast Cells/immunology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Proto-Oncogene Proteins c-kit/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/immunology , Salmonella typhimurium/isolation & purification , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Spleen/pathology , Spleen/ultrastructure , Survival Rate
16.
Blood ; 106(2): 584-92, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15811956

ABSTRACT

In vivo, leukocyte transendothelial migration (TEM) occurs at endothelial cell junctions (paracellular) and nonjunctional (transcellular) locations, whereas in vitro models report that TEM is mostly paracellular. The mechanisms that control the route of leukocyte TEM remain unknown. Here we tested the hypothesis that elevated intercellular adhesion molecule-1 (ICAM-1) expression regulates the location of polymorphonuclear leukocyte (PMN) TEM. We used an in vitro flow model of tumor necrosis factor-alpha (TNF-alpha)-activated human umbilical vein endothelium cells (HUVECs) or an HUVEC cell line transfected with ICAM-1GFP (green fluorescent protein) and live-cell fluorescence microscopy to quantify the location of PMN adhesion and TEM. We observed robust transcellular TEM with TNF-alpha-activated HUVECs and ICAM-1GFP immortalized HUVECS (iHUVECs). In contrast, primary CD3+ T lymphocytes exclusively used a paracellular route. Endothelial ICAM-1 was identified as essential for both paracellular and transcellular PMN transmigration, and interfering with ICAM-1 cytoplasmic tail function preferentially reduced transcellular TEM. We also found that ICAM-1 surface density and distribution as well as endothelial cell shape contributed to transcellular TEM. In summary, ICAM-1 promotes junctional and nonjunctional TEM across inflamed vascular endothelium via distinct cytoplasmic tail associations.


Subject(s)
Intercellular Adhesion Molecule-1/physiology , Neutrophils/physiology , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Sequence , Base Sequence , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , In Vitro Techniques , Intercellular Adhesion Molecule-1/chemistry , Intercellular Adhesion Molecule-1/genetics , Neutrophils/cytology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/pharmacology , T-Lymphocytes/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...