Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Cancer ; 4(10): 1474-1490, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37783807

ABSTRACT

Acute myeloid leukemia (AML), the most frequent leukemia in adults, is driven by recurrent somatically acquired genetic lesions in a restricted number of genes. Treatment with tyrosine kinase inhibitors has demonstrated that targeting of prevalent FMS-related receptor tyrosine kinase 3 (FLT3) gain-of-function mutations can provide significant survival benefits for patients, although the efficacy of FLT3 inhibitors in eliminating FLT3-mutated clones is variable. We identified a T cell receptor (TCR) reactive to the recurrent D835Y driver mutation in the FLT3 tyrosine kinase domain (TCRFLT3D/Y). TCRFLT3D/Y-redirected T cells selectively eliminated primary human AML cells harboring the FLT3D835Y mutation in vitro and in vivo. TCRFLT3D/Y cells rejected both CD34+ and CD34- AML in mice engrafted with primary leukemia from patients, reaching minimal residual disease-negative levels, and eliminated primary CD34+ AML leukemia-propagating cells in vivo. Thus, T cells targeting a single shared mutation can provide efficient immunotherapy toward selective elimination of clonally involved primary AML cells in vivo.


Subject(s)
Leukemia, Myeloid, Acute , Protein-Tyrosine Kinases , Adult , Humans , Animals , Mice , Mutation , Protein-Tyrosine Kinases/genetics , Gain of Function Mutation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell/genetics , fms-Like Tyrosine Kinase 3/genetics
2.
Stud Health Technol Inform ; 282: 259-270, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34085974

ABSTRACT

The purpose of this study was to review the qualitative literature on cognitive accessibility in a digital environment and areas of inquiry for future qualitative research in this context. The focus of this literature review was to identify qualitative research in the cognitive accessibility field and how commonly this term is mentioned in qualitative research articles. In this study, a literature review was conducted on selected qualitative research studies performed globally related to cognitive accessibility. This literature review analysed through meta-synthesis. Based on the results of the literature review, an understanding of existing qualitative research was obtained in the cognitive accessibility field, as well as topics for further qualitative research in the cognitive accessibility field.


Subject(s)
Cognition , Qualitative Research
3.
Sci Rep ; 7(1): 4916, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687807

ABSTRACT

The collagen- and calcium-binding EGF domains 1 (CCBE1) protein is necessary for lymphangiogenesis. Its C-terminal collagen-like domain was shown to be required for the activation of the major lymphangiogenic growth factor VEGF-C (Vascular Endothelial Growth Factor-C) along with the ADAMTS3 (A Disintegrin And Metalloproteinase with Thrombospondin Motifs-3) protease. However, it remained unclear how the N-terminal domain of CCBE1 contributed to lymphangiogenic signaling. Here, we show that efficient activation of VEGF-C requires its C-terminal domain both in vitro and in a transgenic mouse model. The N-terminal EGF-like domain of CCBE1 increased VEGFR-3 signaling by colocalizing pro-VEGF-C with its activating protease to the lymphatic endothelial cell surface. When the ADAMTS3 amounts were limited, proteolytic activation of pro-VEGF-C was supported by the N-terminal domain of CCBE1, but not by its C-terminal domain. A single amino acid substitution in ADAMTS3, identified from a lymphedema patient, was associated with abnormal CCBE1 localization. These results show that CCBE1 promotes VEGFR-3 signaling and lymphangiogenesis by different mechanisms, which are mediated independently by the two domains of CCBE1: by enhancing the cleavage activity of ADAMTS3 and by facilitating the colocalization of VEGF-C and ADAMTS3. These new insights should be valuable in developing new strategies to therapeutically target VEGF-C/VEGFR-3-induced lymphangiogenesis.


Subject(s)
Calcium-Binding Proteins/genetics , Endothelial Cells/metabolism , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Lymphedema/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , COS Cells , Calcium-Binding Proteins/metabolism , Chlorocebus aethiops , Endothelial Cells/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Lymphatic Vessels/pathology , Lymphedema/metabolism , Lymphedema/pathology , Mice , Mice, Transgenic , NIH 3T3 Cells , Protein Domains , Protein Precursors/genetics , Protein Precursors/metabolism , Proteolysis , Signal Transduction , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
4.
Circ Res ; 120(8): 1263-1275, 2017 Apr 14.
Article in English | MEDLINE | ID: mdl-28179432

ABSTRACT

RATIONALE: Lymphatic vessel formation and function constitutes a physiologically and pathophysiologically important process, but its genetic control is not well understood. OBJECTIVE: Here, we identify the secreted Polydom/Svep1 protein as essential for the formation of the lymphatic vasculature. We analyzed mutants in mice and zebrafish to gain insight into the role of Polydom/Svep1 in the lymphangiogenic process. METHODS AND RESULTS: Phenotypic analysis of zebrafish polydom/svep1 mutants showed a decrease in venous and lymphovenous sprouting, which leads to an increased number of intersegmental arteries. A reduced number of primordial lymphatic cells populated the horizontal myoseptum region but failed to migrate dorsally or ventrally, resulting in severe reduction of the lymphatic trunk vasculature. Corresponding mutants in the mouse Polydom/Svep1 gene showed normal egression of Prox-1+ cells from the cardinal vein at E10.5, but at E12.5, the tight association between the cardinal vein and lymphatic endothelial cells at the first lymphovenous contact site was abnormal. Furthermore, mesenteric lymphatic structures at E18.5 failed to undergo remodeling events in mutants and lacked lymphatic valves. In both fish and mouse embryos, the expression of the gene suggests a nonendothelial and noncell autonomous mechanism. CONCLUSIONS: Our data identify zebrafish and mouse Polydom/Svep1 as essential extracellular factors for lymphangiogenesis. Expression of the respective genes by mesenchymal cells in intimate proximity with venous and lymphatic endothelial cells is required for sprouting and migratory events in zebrafish and for remodeling events of the lymphatic intraluminal valves in mouse embryos.


Subject(s)
Endothelial Cells/metabolism , Evolution, Molecular , Lymphangiogenesis , Lymphatic Vessels/metabolism , Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Calcium-Binding Proteins , Cell Adhesion Molecules , Cell Communication , Cell Movement , Endothelial Cells/pathology , Endothelium, Lymphatic/abnormalities , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/physiopathology , Gene Expression Regulation, Developmental , Genotype , Lymphatic Vessels/abnormalities , Lymphatic Vessels/physiopathology , Mesoderm/metabolism , Mutation , Phenotype , Proteins/genetics , Signal Transduction , Time Factors , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Front Immunol ; 8: 1718, 2017.
Article in English | MEDLINE | ID: mdl-29321773

ABSTRACT

T cells can recognize peptides encoded by mutated genes, but analysis of tumor-infiltrating lymphocytes suggests that very few neoantigens spontaneously elicit T-cell responses. This may be an important reason why immune checkpoint inhibitors are mainly effective in tumors with a high mutational burden. Reasons for clinically insufficient responses to neoantigens might be inefficient priming, inhibition, or deletion of the cognate T cells. Responses can be dramatically improved by cancer immunotherapy such as checkpoint inhibition, but often with temporary effects. By contrast, T cells from human leukocyte antigen (HLA)-matched donors can cure diseases such as chronic myeloid leukemia. The therapeutic effect is mediated by donor T cells recognizing polymorphic peptides for which the donor and patient are disparate, presented on self-HLA. Donor T-cell repertoires are unbiased by the immunosuppressive environment of the tumor. A recent study demonstrated that T cells from healthy individuals are able to respond to neoantigens that are ignored by tumor-infiltrating T cells of melanoma patients. In this review, we discuss possible reasons why neoantigens escape host T cells and how these limitations may be overcome by utilization of donor-derived T-cell repertoires to facilitate rational design of neoantigen-targeted immunotherapy.

6.
Mol Oncol ; 9(10): 2019-42, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26548533

ABSTRACT

T lymphocytes can be redirected to recognize a tumor target and harnessed to combat cancer by genetic introduction of T-cell receptors of a defined specificity. This approach has recently mediated encouraging clinical responses in patients with cancers previously regarded as incurable. However, despite the great promise, T-cell receptor gene therapy still faces a multitude of obstacles. Identification of epitopes that enable effective targeting of all the cells in a heterogeneous tumor while sparing normal tissues remains perhaps the most demanding challenge. Experience from clinical trials has revealed the dangers associated with T-cell receptor gene therapy and highlighted the need for reliable preclinical methods to identify potentially hazardous recognition of both intended and unintended epitopes in healthy tissues. Procedures for manufacturing large and highly potent T-cell populations can be optimized to enhance their antitumor efficacy. Here, we review the current knowledge gained from preclinical models and clinical trials using adoptive transfer of T-cell receptor-engineered T lymphocytes, discuss the major challenges involved and highlight potential strategies to increase the safety and efficacy to make T-cell receptor gene therapy a standard-of-care for large patient groups.


Subject(s)
Genetic Therapy , Receptors, Antigen, T-Cell/genetics , Animals , Humans , Lymphocyte Depletion , Mice
7.
Circulation ; 129(19): 1962-71, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24552833

ABSTRACT

BACKGROUND: Hennekam lymphangiectasia-lymphedema syndrome (Online Mendelian Inheritance in Man 235510) is a rare autosomal recessive disease, which is associated with mutations in the CCBE1 gene. Because of the striking phenotypic similarity of embryos lacking either the Ccbe1 gene or the lymphangiogenic growth factor Vegfc gene, we searched for collagen- and calcium-binding epidermal growth factor domains 1 (CCBE1) interactions with the vascular endothelial growth factor-C (VEGF-C) growth factor signaling pathway, which is critical in embryonic and adult lymphangiogenesis. METHODS AND RESULTS: By analyzing VEGF-C produced by CCBE1-transfected cells, we found that, whereas CCBE1 itself does not process VEGF-C, it promotes proteolytic cleavage of the otherwise poorly active 29/31-kDa form of VEGF-C by the A disintegrin and metalloprotease with thrombospondin motifs-3 protease, resulting in the mature 21/23-kDa form of VEGF-C, which induces increased VEGF-C receptor signaling. Adeno-associated viral vector-mediated transduction of CCBE1 into mouse skeletal muscle enhanced lymphangiogenesis and angiogenesis induced by adeno-associated viral vector-VEGF-C. CONCLUSIONS: These results identify A disintegrin and metalloprotease with thrombospondin motifs-3 as a VEGF-C-activating protease and reveal a novel type of regulation of a vascular growth factor by a protein that enhances its proteolytic cleavage and activation. The results suggest that CCBE1 is a potential therapeutic tool for the modulation of lymphangiogenesis and angiogenesis in a variety of diseases that involve the lymphatic system, such as lymphedema or lymphatic metastasis.


Subject(s)
ADAM Proteins/metabolism , Calcium-Binding Proteins/metabolism , Lymphangiogenesis/physiology , Procollagen N-Endopeptidase/metabolism , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor C/metabolism , ADAMTS Proteins , Adenoviridae/genetics , Animals , Calcium-Binding Proteins/genetics , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred Strains , Models, Animal , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/physiology , Transfection , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
8.
Development ; 141(6): 1239-49, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24523457

ABSTRACT

The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.


Subject(s)
Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , DNA/genetics , Gene Expression Regulation, Developmental , Humans , Lymphangiogenesis/genetics , MAP Kinase Signaling System , Mice , Molecular Sequence Data , Point Mutation , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Development ; 140(13): 2776-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23698350

ABSTRACT

The formation and lumenization of blood vessels has been studied in some detail, but there is little understanding of the morphogenetic mechanisms by which endothelial cells (ECs) forming large caliber vessels aggregate, align themselves and finally form a lumen that can support blood flow. Here, we focus on the development of the zebrafish common cardinal veins (CCVs), which collect all the blood from the embryo and transport it back to the heart. We show that the angioblasts that eventually form the definitive CCVs become specified as a separate population distinct from the angioblasts that form the lateral dorsal aortae. The subsequent development of the CCVs represents a novel mechanism of vessel formation, during which the ECs delaminate and align along the inner surface of an existing luminal space. Thereby, the CCVs are initially established as open-ended endothelial tubes, which extend as single EC sheets along the flow routes of the circulating blood and eventually enclose the entire lumen in a process that we term 'lumen ensheathment'. Furthermore, we found that the initial delamination of the ECs as well as the directional migration within the EC sheet depend on Cadherin 5 function. By contrast, EC proliferation within the growing CCV is controlled by Vascular endothelial growth factor C, which is provided by circulating erythrocytes. Our findings not only identify a novel mechanism of vascular lumen formation, but also suggest a new form of developmental crosstalk between hematopoietic and endothelial cell lineages.


Subject(s)
Embryo, Nonmammalian/metabolism , Veins/embryology , Veins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Movement/genetics , Cell Movement/physiology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Methods Cell Biol ; 105: 223-38, 2011.
Article in English | MEDLINE | ID: mdl-21951532

ABSTRACT

The mammalian lymphatic vasculature has an important function in the maintenance of tissue fluid homeostasis, absorption of dietary lipids, and immune surveillance. The lymphatic vessels are also recruited by many tumors as primary routes for metastasis and mediate immune responses in inflammatory diseases, whereas dysfunction of the lymphatic drainage leads to lymphedema. The characterization of a lymphatic vasculature in zebrafish has made the advantages of this small model organism, the suitability for intravital time-lapse imaging of developmental processes and the amenability for chemical and forward genetic screens, available to lymphatic vascular research. Here we review our current understanding of embryonic lymphangiogenesis in zebrafish, its molecular and anatomical similarities to mammalian lymphatic vascular development, and the possibilities zebrafish offers to complement mouse models and cell culture assays in the lymphangiogenesis field.


Subject(s)
Developmental Biology/methods , Lymphangiogenesis/physiology , Lymphatic Diseases/pathology , Lymphatic Vessels/pathology , Lymphography/methods , Molecular Imaging/methods , Signal Transduction/physiology , Time-Lapse Imaging/methods , Zebrafish , Animals , Disease Models, Animal , Fluorescent Dyes/analysis , Humans , Lipid Metabolism , Lymphatic Diseases/genetics , Lymphatic Vessels/embryology , Mice , Zebrafish/embryology , Zebrafish/physiology
11.
Circ Res ; 109(5): 486-91, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21778431

ABSTRACT

RATIONALE: Collagen- and calcium-binding EGF domains 1 (CCBE1) has been associated with Hennekam syndrome, in which patients have lymphedema, lymphangiectasias, and other cardiovascular anomalies. Insight into the molecular role of CCBE1 is completely lacking, and mouse models for the disease do not exist. OBJECTIVE: CCBE1 deficient mice were generated to understand the function of CCBE1 in cardiovascular development, and CCBE1 recombinant protein was used in both in vivo and in vitro settings to gain insight into the molecular function of CCBE1. METHODS AND RESULTS: Phenotypic analysis of murine Ccbe1 mutant embryos showed a complete lack of definitive lymphatic structures, even though Prox1(+) lymphatic endothelial cells get specified within the cardinal vein. Mutant mice die prenatally. Proximity ligation assays indicate that vascular endothelial growth factor receptor 3 activation appears unaltered in mutants. Human CCBE1 protein binds to components of the extracellular matrix in vitro, and CCBE1 protein strongly enhances vascular endothelial growth factor-C-mediated lymphangiogenesis in a corneal micropocket assay. CONCLUSIONS: Our data identify CCBE1 as a factor critically required for budding and migration of Prox-1(+) lymphatic endothelial cells from the cardinal vein. CCBE1 probably exerts these effects through binding to components of the extracellular matrix. CCBE1 has little lymphangiogenic effect on its own but dramatically enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo. Thus, our data suggest CCBE1 to be essential but not sufficient for lymphangiogenesis.


Subject(s)
Calcium-Binding Proteins/physiology , Endothelium, Lymphatic/blood supply , Endothelium, Lymphatic/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Lymphatic Vessels/metabolism , Tumor Suppressor Proteins/physiology , Vascular Endothelial Growth Factor C/metabolism , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cornea/blood supply , Cornea/cytology , Cornea/metabolism , Endothelium, Lymphatic/cytology , Humans , Lymphangiogenesis/genetics , Mice , Mice, Knockout , Protein Binding/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/physiology
12.
Blood ; 118(4): 1154-62, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21566091

ABSTRACT

Notch signaling plays a central role in cell-fate determination, and its role in lateral inhibition in angiogenic sprouting is well established. However, the role of Notch signaling in lymphangiogenesis, the growth of lymphatic vessels, is poorly understood. Here we demonstrate Notch pathway activity in lymphatic endothelial cells (LECs), as well as induction of delta-like ligand 4 (Dll4) and Notch target genes on stimulation with VEGF or VEGF-C. Suppression of Notch signaling by a soluble form of Dll4 (Dll4-Fc) synergized with VEGF in inducing LEC sprouting in 3-dimensional (3D) fibrin gel assays. Expression of Dll4-Fc in adult mouse ears promoted lymphangiogenesis, which was augmented by coexpressing VEGF. Lymphangiogenesis triggered by Notch inhibition was suppressed by a monoclonal VEGFR-2 Ab as well as soluble VEGF and VEGF-C/VEGF-D ligand traps. LECs transduced with Dll4 preferentially adopted the tip cell position over nontransduced cells in 3D sprouting assays, suggesting an analogous role for Dll4/Notch in lymphatic and blood vessel sprouting. These results indicate that the Notch pathway controls lymphatic endothelial quiescence, and explain why LECs are poorly responsive to VEGF compared with VEGF-C. Understanding the role of the Notch pathway in lymphangiogenesis provides further insight for the therapeutic manipulation of the lymphatic vessels.


Subject(s)
Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Mol Cell Biol ; 30(14): 3620-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20479124

ABSTRACT

The lymphatic vascular system maintains tissue fluid homeostasis, helps mediate afferent immune responses, and promotes cancer metastasis. To address the role microRNAs (miRNAs) play in the development and function of the lymphatic vascular system, we defined the in vitro miRNA expression profiles of primary human lymphatic endothelial cells (LECs) and blood vascular endothelial cells (BVECs) and identified four BVEC signature and two LEC signature miRNAs. Their vascular lineage-specific expression patterns were confirmed in vivo by quantitative real-time PCR and in situ hybridization. Functional characterization of the BVEC signature miRNA miR-31 identified a novel BVEC-specific posttranscriptional regulatory mechanism that inhibits the expression of lymphatic lineage-specific transcripts in vitro. We demonstrate that suppression of lymphatic differentiation is partially mediated via direct repression of PROX1, a transcription factor that functions as a master regulator of lymphatic lineage-specific differentiation. Finally, in vivo studies of Xenopus and zebrafish demonstrated that gain of miR-31 function impaired venous sprouting and lymphatic vascular development, thus highlighting the importance of miR-31 as a negative regulator of lymphatic development. Collectively, our findings identify miR-31 is a potent regulator of vascular lineage-specific differentiation and development in vertebrates.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Lymphatic System/cytology , Lymphatic System/growth & development , MicroRNAs/genetics , Animals , Animals, Genetically Modified , Base Sequence , Blood Vessels/cytology , Blood Vessels/growth & development , Blood Vessels/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization , In Vitro Techniques , Lymphatic System/metabolism , Mice , MicroRNAs/metabolism , Polymerase Chain Reaction , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
14.
Circulation ; 121(12): 1413-22, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231530

ABSTRACT

BACKGROUND: Lymphatic network and chemokine-mediated signals are essential for leukocyte traffic during the proximal steps of alloimmune response. We aimed to determine the role of lymphatic vessels and their principal growth signaling pathway, vascular endothelial growth factor (VEGF)-C/D/VEGFR-3, during acute and chronic rejection in cardiac allografts. METHODS AND RESULTS: Analysis of heterotopically transplanted rat cardiac allografts showed that chronic rejection increased VEGF-C(+) inflammatory cell and hyaluronan receptor-1 (LYVE-1)(+) lymphatic vessel density. Allograft lymphatic vessels were VEGFR-3(+), contained antigen-presenting cells, and produced dendritic cell chemokine CCL21. Experiments with VEGFR-3/LacZ mice or mice with green fluorescent protein-positive bone marrow cells as cardiac allograft recipients showed that allograft lymphatic vessels originated almost exclusively from donor cells. Intraportal adenoviral VEGFR-3-Ig (Ad.VEGFR-3-Ig/VEGF-C/D-Trap) perfusion was used to inhibit VEGF-C/D/VEGFR-3 signaling. Recipient treatment with Ad.VEGFR-3-Ig prolonged rat cardiac allograft survival. Ad.VEGFR-3-Ig did not affect allograft lymphangiogenesis but was linked to reduced CCL21 production and CD8(+) effector cell entry in the allograft. Concomitantly, Ad.VEGFR-3-Ig reduced OX62(+) dendritic cell recruitment and increased transcription factor Foxp3 expression in the spleen. In separate experiments, treatment with a neutralizing monoclonal VEGFR-3 antibody reduced arteriosclerosis, the number of activated lymphatic vessels expressing VEGFR-3 and CCL21, and graft-infiltrating CD4(+) T cells in chronically rejecting mouse cardiac allografts. CONCLUSIONS: These results show that VEGFR-3 participates in immune cell traffic from peripheral tissues to secondary lymphoid organs by regulating allograft lymphatic vessel CCL21 production and suggest VEGFR-3 inhibition as a novel lymphatic vessel-targeted immunomodulatory therapy for cardiac allograft rejection and arteriosclerosis.


Subject(s)
Arteriosclerosis/prevention & control , Chemokine CCL21/biosynthesis , Heart Transplantation/immunology , Immunomodulation/drug effects , Lymphatic Vessels/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigen-Presenting Cells , Arteriosclerosis/drug therapy , Cell Movement/immunology , Graft Rejection/immunology , Mice , Mice, Knockout , Rats , Signal Transduction/immunology , Vascular Endothelial Growth Factor Receptor-3/immunology
15.
Circ Res ; 103(9): 1018-26, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18757827

ABSTRACT

Vascular endothelial growth factor (VEGF)-B is poorly angiogenic but prominently expressed in metabolically highly active tissues, including the heart. We produced mice expressing a cardiac-specific VEGF-B transgene via the alpha-myosin heavy chain promoter. Surprisingly, the hearts of the VEGF-B transgenic mice showed concentric cardiac hypertrophy without significant changes in heart function. The cardiac hypertrophy was attributable to an increased size of the cardiomyocytes. Blood capillary size was increased, whereas the number of blood vessels per cell nucleus remained unchanged. Despite the cardiac hypertrophy, the transgenic mice had lower heart rate and blood pressure than their littermates, and they responded similarly to angiotensin II-induced hypertension, confirming that the hypertrophy does not compromise heart function. Interestingly, the isolated transgenic hearts had less cardiomyocyte damage after ischemia. Significantly increased ceramide and decreased triglyceride levels were found in the transgenic hearts. This was associated with structural changes and eventual lysis of mitochondria, resulting in accumulation of intracellular vacuoles in cardiomyocytes and increased death of the transgenic mice, apparently because of mitochondrial lipotoxicity in the heart. These results suggest that VEGF-B regulates lipid metabolism, an unexpected function for an angiogenic growth factor.


Subject(s)
Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Lipid Metabolism , Myocardium/metabolism , Vascular Endothelial Growth Factor B/metabolism , Ventricular Function, Left , Angiotensin II , Animals , Blood Pressure , Capillaries/metabolism , Capillaries/pathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Size , Ceramides/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Heart Rate , Humans , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Neovascularization, Physiologic , Oxidation-Reduction , Promoter Regions, Genetic , Skin/blood supply , Skin/metabolism , Time Factors , Triglycerides/metabolism , Up-Regulation , Vascular Endothelial Growth Factor B/genetics , Ventricular Myosins/genetics
16.
Blood ; 112(5): 1547-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18725566
17.
Annu Rev Pathol ; 3: 367-97, 2008.
Article in English | MEDLINE | ID: mdl-18039141

ABSTRACT

The lymphatic vasculature is essential for the maintenance of tissue fluid balance, immune surveillance, and adsorption fatty acids in the gut. The lymphatic vessels are also crucially involved in the pathogenesis of diseases such as tumor metastasis, lymphedema, and various inflammatory conditions. Attempts to control or treat these diseases have drawn a lot of interest to lymphatic vascular research during the past few years. Recently, several markers specific for lymphatic endothelium and models for lymphatic vascular research have been characterized, enabling great technical progress in lymphatic vascular biology, and many critical regulators of lymphatic vessel growth have been identified. Despite these significant achievements, our understanding of the lymphatic vessel development and pathogenesis is still rather limited. Several key questions remain to be resolved, including the relative contributions of different pathways targeting lymphatic vasculature, the molecular and cellular processes of lymphatic maturation, and the detailed mechanisms of tumor metastasis via the lymphatic system.


Subject(s)
Gene Expression Regulation, Developmental , Lymphangiogenesis/genetics , Lymphatic Vessels/pathology , Lymphedema/pathology , Animals , Biomarkers/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Humans , Lymphatic Vessels/metabolism , Lymphedema/genetics , Lymphedema/metabolism , Mice , Models, Animal , Molecular Biology/methods , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
18.
Circ Res ; 100(10): 1460-7, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17478734

ABSTRACT

Vascular endothelial growth factor (VEGF)-C and VEGF-D require proteolytic cleavage of the carboxy terminal silk-homology domain for activation. To study the functions of the VEGF-C propeptides, we engineered a chimeric growth factor protein, VEGF-CAC, composed of the amino- and carboxy-terminal propeptides of VEGF-C fused to the receptor-activating core domain of VEGF. Like VEGF-C, VEGF-CAC underwent proteolytic cleavage, and like VEGF, it bound to and activated VEGF receptor-1 and VEGF receptor-2, but not the VEGF-C receptor VEGF receptor-3. VEGF-CAC also bound to neuropilins in a heparin-dependent manner. Strikingly, when VEGF-CAC was expressed via an adenovirus vector in the ear skin of immunodeficient mice, it proved to be a more potent inducer of capillary angiogenesis than VEGF. The VEGF-CAC-induced vessels differed greatly from those induced by VEGF, as they formed a very dense and fine network of pericyte and basement membrane-covered capillaries that were functional, as shown by lectin perfusion experiments. VEGF-CAC could prove useful in proangiogenic therapies in patients experiencing tissue ischemia.


Subject(s)
Neovascularization, Physiologic/drug effects , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor C/pharmacology , Adenoviridae/genetics , Animals , Basement Membrane/drug effects , Capillaries/drug effects , Capillaries/physiology , Cells, Cultured , Humans , Lymphatic Vessels/drug effects , Lymphatic Vessels/physiology , Mice , Pericytes/drug effects , Protein Structure, Tertiary , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/chemistry , Vascular Endothelial Growth Factor C/metabolism
19.
Am J Pathol ; 169(2): 708-18, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877368

ABSTRACT

Lymphatic vessel plasticity and stability are of considerable importance when attempting to treat diseases associated with the lymphatic vasculature. Development of lymphatic vessels during embryogenesis is dependent on vascular endothelial growth factor (VEGF)-C but not VEGF-D. Using a recombinant adenovirus encoding a soluble form of their receptor VEGFR-3 (AdVEGFR-3-Ig), we studied lymphatic vessel dependency on VEGF-C and VEGF-D induced VEGFR-3 signaling in postnatal and adult mice. Transduction with AdVEGFR-3-Ig led to regression of lymphatic capillaries and medium-sized lymphatic vessels in mice under 2 weeks of age without affecting collecting lymphatic vessels or the blood vasculature. No effect was observed after this period. The lymphatic capillaries of neonatal mice also regressed partially in response to recombinant VEGFR-3-Ig or blocking antibodies against VEGFR-3, but not to adenovirus-encoded VEGFR-2-Ig. Despite sustained inhibitory VEGFR-3-Ig levels, lymphatic vessel regrowth was observed at 4 weeks of age. Interestingly, whereas transgenic expression of VEGF-C in the skin induced lymphatic hyperplasia even during embryogenesis, similar expression of VEGF-D resulted in lymphangiogenesis predominantly after birth. These results indicate considerable plasticity of lymphatic vessels during the early postnatal period but not thereafter, suggesting that anti-lymphangiogenic therapy can be safely applied in adults.


Subject(s)
Growth Substances/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Adenoviridae/genetics , Animals , Animals, Newborn , Humans , Hyperplasia , Ligands , Lymphatic Vessels/pathology , Lymphatic Vessels/physiology , Mice , Regeneration , Solubility , Transgenes , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor D/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/blood , Vascular Endothelial Growth Factor Receptor-3/immunology
20.
FASEB J ; 20(9): 1462-72, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816121

ABSTRACT

Lymphatic vascular development is regulated by vascular endothelial growth factor receptor-3 (VEGFR-3), which is activated by its ligands VEGF-C and VEGF-D. Neuropilin-2 (NP2), known to be involved in neuronal development, has also been implicated to play a role in lymphangiogenesis. We aimed to elucidate the mechanism by which NP2 is involved in lymphatic endothelial cell signaling. By in vitro binding studies we found that both VEGF-C and VEGF-D interact with NP2, VEGF-C in a heparin-independent and VEGF-D in a heparin-dependent manner. We also mapped the domains of VEGF-C and NP2 required for their binding. The functional importance of the interaction of NP2 with the lymphangiogenic growth factors was demonstrated by cointernalization of NP2 along with VEGFR-3 in endocytic vesicles of lymphatic endothelial cells upon stimulation with VEGF-C or VEGF-D. NP2 also interacted with VEGFR-3 in coprecipitation studies. Our results show that NP2 is directly involved in an active signaling complex with the key regulators of lymphangiogenesis and thus suggest a mechanism by which NP2 functions in the development of the lymphatic vasculature.


Subject(s)
Neovascularization, Physiologic , Neuropilin-2/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers , Drosophila , Endothelium, Vascular/physiology , Heparin/physiology , Humans , Kidney , Lymphatic System/physiology , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Signal Transduction , Swine , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor D/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...