Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Ther ; 29(9): 2841-2853, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33940155

ABSTRACT

A primary challenge in lentiviral gene therapy of ß-hemoglobinopathies is to maintain low vector copy numbers to avoid genotoxicity while being reliably therapeutic for all genotypes. We designed a high-titer lentiviral vector, LVß-shα2, that allows coordinated expression of the therapeutic ßA-T87Q-globin gene and of an intron-embedded miR-30-based short hairpin RNA (shRNA) selectively targeting the α2-globin mRNA. Our approach was guided by the knowledge that moderate reduction of α-globin chain synthesis ameliorates disease severity in ß-thalassemia. We demonstrate that LVß-shα2 reduces α2-globin mRNA expression in erythroid cells while keeping α1-globin mRNA levels unchanged and ßA-T87Q-globin gene expression identical to the parent vector. Compared with the first ßA-T87Q-globin lentiviral vector that has received conditional marketing authorization, BB305, LVß-shα2 shows 1.7-fold greater potency to improve α/ß ratios. It may thus result in greater therapeutic efficacy and reliability for the most severe types of ß-thalassemia and provide an improved benefit/risk ratio regardless of the ß-thalassemia genotype.


Subject(s)
Genetic Vectors/administration & dosage , RNA, Small Interfering/genetics , alpha-Globins/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , Cell Line , Cells, Cultured , Down-Regulation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Genotype , Humans , K562 Cells , Lentivirus/genetics , Lentivirus/physiology , MicroRNAs/antagonists & inhibitors , Primary Cell Culture , Viral Load , beta-Thalassemia/therapy
2.
Sci Rep ; 8(1): 5482, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615807

ABSTRACT

Alternate splicing is a critical regulator of gene expression in eukaryotes, however genetic mutations can cause erroneous splicing and disease. Most recorded splicing disorders are caused by mutations of splice donor/acceptor sites, however intronic mutations can affect splicing. Clinical exome analyses largely ignore intronic sequence, limiting the detection of mutations to within coding regions. We describe 'Trooper', a novel mouse model of CHARGE syndrome harbouring a pathogenic point mutation in Chd7. The mutation is 18 nucleotides upstream of exon 10 and creates a cryptic acceptor site, causing exon skipping and partial intron retention. This mutation, though detectable in exome sequence, was initially dismissed by computational filtering due to its intronic location. The Trooper strain exhibited many of the previously described CHARGE-like anomalies of CHD7 deficient mouse lines; including hearing impairment, vestibular hypoplasia and growth retardation. However, more common features such as facial asymmetry and circling were rarely observed. Recognition of these characteristic features prompted manual reexamination of Chd7 sequence and subsequent validation of the intronic mutation, highlighting the importance of phenotyping alongside exome analyses. The Trooper mouse serves as a valuable model of atypical CHARGE syndrome and reveals a molecular mechanism that may underpin milder clinical presentation of the syndrome.


Subject(s)
CHARGE Syndrome/genetics , DNA-Binding Proteins/genetics , Introns/genetics , Mutation , RNA Splice Sites/genetics , RNA Splicing/genetics , Animals , Base Sequence , CHARGE Syndrome/physiopathology , Disease Models, Animal , Hearing/genetics , Male , Mice , Mice, Inbred BALB C , Phenotype
3.
Blood ; 129(23): 3087-3099, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28325862

ABSTRACT

ß-Thalassemia is associated with several abnormalities of the innate immune system. Neutrophils in particular are defective, predisposing patients to life-threatening bacterial infections. The molecular and cellular mechanisms involved in impaired neutrophil function remain incompletely defined. We used the Hbbth3/+ ß-thalassemia mouse and hemoglobin E (HbE)/ß-thalassemia patients to investigate dysregulated neutrophil activity. Mature neutrophils from Hbbth3/+ mice displayed a significant reduction in chemotaxis, opsonophagocytosis, and production of reactive oxygen species, closely mimicking the defective immune functions observed in ß-thalassemia patients. In Hbbth3/+ mice, the expression of neutrophil CXCR2, CD11b, and reduced NAD phosphate oxidase components (p22phox, p67phox, and gp91phox) were significantly reduced. Morphological analysis of Hbbth3/+ neutrophils showed that a large percentage of mature phenotype neutrophils (Ly6GhiLy6Clow) appeared as band form cells, and a striking expansion of immature (Ly6GlowLy6Clow) hyposegmented neutrophils, consisting mainly of myelocytes and metamyelocytes, was noted. Intriguingly, expression of an essential mediator of neutrophil terminal differentiation, the ets transcription factor PU.1, was significantly decreased in Hbbth3/+ neutrophils. In addition, in vivo infection with Streptococcus pneumoniae failed to induce PU.1 expression or upregulate neutrophil effector functions in Hbbth3/+ mice. Similar changes to neutrophil morphology and PU.1 expression were observed in splenectomized and nonsplenectomized HbE/ß-thalassemia patients. This study provides a mechanistic insight into defective neutrophil maturation in ß-thalassemia patients, which contributes to deficiencies in neutrophil effector functions.


Subject(s)
Neutrophils/immunology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , beta-Thalassemia/genetics , beta-Thalassemia/immunology , Adult , Animals , CD11b Antigen/metabolism , Case-Control Studies , Cell Differentiation/genetics , Cell Differentiation/immunology , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Middle Aged , Neutrophil Activation , Neutrophils/metabolism , Neutrophils/pathology , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/immunology , Reactive Oxygen Species/metabolism , Receptors, Interleukin-8B/metabolism , Trans-Activators/deficiency , Trans-Activators/immunology , Young Adult , beta-Thalassemia/pathology
4.
Biochim Biophys Acta Gene Regul Mech ; 1860(4): 393-404, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28161275

ABSTRACT

During development, the α- and ß-globin genes exhibit a highly conserved pattern of expression, giving rise to several developmental stage-specific hemoglobin variants. Networks of regulatory proteins interact with epigenetic complexes to regulate DNA accessibility and histone modifications, thereby determining appropriate patterns of globin gene expression. In this review, we focus on recent advances in the understanding of the molecular mechanisms that underpin globin gene expression, focusing on multi-subunit regulatory complexes that bind to specific regions of DNA to orchestrate globin gene transcription throughout development.


Subject(s)
Epigenesis, Genetic , Genetic Loci , beta-Globins/genetics , Animals , Chromatin/metabolism , Histones/metabolism , Humans , Protein Processing, Post-Translational/genetics
5.
J Blood Med ; 7: 263-274, 2016.
Article in English | MEDLINE | ID: mdl-27853395

ABSTRACT

The structural and functional conservation of hemoglobin throughout mammals has made the laboratory mouse an exceptionally useful organism in which to study both the protein and the individual globin genes. Early researchers looked to the globin genes as an excellent model in which to examine gene regulation - bountifully expressed and displaying a remarkably consistent pattern of developmental activation and silencing. In parallel with the growth of research into expression of the globin genes, mutations within the ß-globin gene were identified as the cause of the ß-hemoglobinopathies such as sickle cell disease and ß-thalassemia. These lines of enquiry stimulated the development of transgenic mouse models, first carrying individual human globin genes and then substantial human genomic fragments incorporating the multigenic human ß-globin locus and regulatory elements. Finally, mice were devised carrying mutant human ß-globin loci on genetic backgrounds deficient in the native mouse globins, resulting in phenotypes of sickle cell disease or ß-thalassemia. These years of work have generated a group of model animals that display many features of the ß-hemoglobinopathies and provided enormous insight into the mechanisms of gene regulation. Substantive differences in the expression of human and mouse globins during development have also come to light, revealing the limitations of the mouse model, but also providing opportunities to further explore the mechanisms of globin gene regulation. In addition, animal models of ß-hemoglobinopathies have demonstrated the feasibility of gene therapy for these conditions, now showing success in human clinical trials. Such models remain in use to dissect the molecular events of globin gene regulation and to identify novel treatments based upon the reactivation of developmentally silenced γ-globin. Here, we describe the development of animal models to investigate globin switching and the ß-hemoglobinopathies, a field that has paralleled the emergence of modern molecular biology and clinical genetics.

6.
Stem Cell Reports ; 7(3): 508-517, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27499201

ABSTRACT

While Cas9 nucleases permit rapid and efficient generation of gene-edited cell lines, the CRISPR-Cas9 system can introduce undesirable "on-target" mutations within the second allele of successfully modified cells via non-homologous end joining (NHEJ). To address this, we fused the Streptococcus pyogenes Cas9 (SpCas9) nuclease to a peptide derived from the human Geminin protein (SpCas9-Gem) to facilitate its degradation during the G1 phase of the cell cycle, when DNA repair by NHEJ predominates. We also use mRNA transfection to facilitate low and transient expression of modified and unmodified versions of Cas9. Although the frequency of homologous recombination was similar for SpCas9-Gem and SpCas9, we observed a marked reduction in the capacity for SpCas9-Gem to induce NHEJ-mediated indels at the target locus. Moreover, in contrast to native SpCas9, we demonstrate that transient SpCas9-Gem expression enables reliable generation of both knockin reporter cell lines and genetically repaired patient-specific induced pluripotent stem cell lines free of unwanted mutations at the targeted locus.


Subject(s)
CRISPR-Cas Systems , Gene Knock-In Techniques , Pluripotent Stem Cells/metabolism , Targeted Gene Repair , Base Sequence , Cell Line , Cellular Reprogramming/genetics , DNA End-Joining Repair , Fibroblasts/cytology , Fibroblasts/metabolism , Geminin/genetics , Geminin/metabolism , Gene Expression , Genes, Reporter , Humans , INDEL Mutation , Pluripotent Stem Cells/cytology , Sequence Analysis, DNA , Transfection
8.
F1000Res ; 42015.
Article in English | MEDLINE | ID: mdl-26918126

ABSTRACT

The rapid advances in the field of genome editing using targeted endonucleases have called considerable attention to the potential of this technology for human gene therapy. Targeted correction of disease-causing mutations could ensure lifelong, tissue-specific expression of the relevant gene, thereby alleviating or resolving a specific disease phenotype. In this review, we aim to explore the potential of this technology for the therapy of ß-thalassemia. This blood disorder is caused by mutations in the gene encoding the ß-globin chain of hemoglobin, leading to severe anemia in affected patients. Curative allogeneic bone marrow transplantation is available only to a small subset of patients, leaving the majority of patients dependent on regular blood transfusions and iron chelation therapy. The transfer of gene-corrected autologous hematopoietic stem cells could provide a therapeutic alternative, as recent results from gene therapy trials using a lentiviral gene addition approach have demonstrated. Genome editing has the potential to further advance this approach as it eliminates the need for semi-randomly integrating viral vectors and their associated risk of insertional mutagenesis. In the following pages we will highlight the advantages and risks of genome editing compared to standard therapy for ß-thalassemia and elaborate on lessons learned from recent gene therapy trials.

9.
Methods Mol Biol ; 1227: 309-21, 2015.
Article in English | MEDLINE | ID: mdl-25239755

ABSTRACT

Gene therapy of inherited diseases requires long-term maintenance of the corrective transgene. Stable integration of the introduced DNA molecule into one of the host cell chromosomes is the simplest strategy for achieving this. However, genotoxicity resulting from random insertion of the transgene raises serious safety concerns that must be addressed if gene therapy is to enter the clinical mainstream. The following method makes use of the Rep integrase of adeno-associated virus to insert a transgene into the human AAVS1 site, a known "safe harbor" region within the human genome. This approach has the potential for application to novel gene therapy strategies for improved safety. In addition, with this method it is also possible to create cell lines carrying BAC transgenes in the AAVS1 site.


Subject(s)
Chromosomes, Artificial, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Dependovirus/metabolism , Genetic Vectors/metabolism , Integrases/metabolism , Viral Proteins/metabolism , Virus Integration/genetics , Base Sequence , Chromosomes, Artificial, Bacterial/genetics , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/chemistry , Humans , Integrases/genetics , Molecular Sequence Data , RNA, Viral/genetics , RNA, Viral/metabolism , Recombination, Genetic , Transfection , Viral Proteins/genetics
10.
Methods Mol Biol ; 1227: 323-43, 2015.
Article in English | MEDLINE | ID: mdl-25239756

ABSTRACT

Bacterial artificial chromosome (BAC) reporter cell lines are generated through stable transfection of a BAC reporter construct wherein the gene of interest is tagged with a reporter gene such as eGFP. The large capacity of BACs (up to 350 kb of genomic sequence) enables the inclusion of all regulatory elements that ensure appropriate regulation of the gene of interest. Furthermore, the reporter gene allows the expression of the gene of interest to be readily detected by flow cytometry. Cell lines can also be easily cultured for extended periods with minimal cost. These features of BAC reporter cell lines make them highly amenable for use in high-throughput screening of large drug libraries for compounds that induce the expression of the gene of interest. This chapter describes a method for generation of BAC reporter cell lines that are suitable as cellular assay systems in high-throughput screening. Briefly, this method involves (A) generation of cell clones stably transfected with a BAC reporter construct, (B) selection of "candidate" cell clones based on the responsiveness to known inducers, (C) confirmation of the integrity of the BAC reporter construct integrated within the candidate clones, and (D) assessment of the developmental regulation of the BAC reporter construct. As an example, we describe the generation of a BAC reporter cell line containing the human ß-globin locus modified to express γ-globin as eGFP for use as a cellular reporter assay for screening of drugs that can reactivate expression of developmentally silenced γ-globin for the treatment of ß-hemoglobin disorders.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Green Fluorescent Proteins/genetics , High-Throughput Screening Assays , Small Molecule Libraries/pharmacology , beta-Globins/genetics , gamma-Globins/genetics , Animals , Chromosomes, Artificial, Bacterial/metabolism , Drug Discovery , Electroporation , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , K562 Cells , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , beta-Globins/metabolism , gamma-Globins/metabolism
11.
FASEB J ; 28(5): 2306-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24443374

ABSTRACT

Expression of fetal γ-globin in adulthood ameliorates symptoms of ß-hemoglobinopathies by compensating for the mutant ß-globin. Reactivation of the silenced γ-globin gene is therefore of substantial clinical interest. To study the regulation of γ-globin expression, we created the GG mice, which carry an intact 183-kb human ß-globin locus modified to express enhanced green fluorescent protein (eGFP) from the Gγ-globin promoter. GG embryos express eGFP first in the yolk sac blood islands and then in the aorta-gonad mesonephros and the fetal liver, the sites of normal embryonic hematopoiesis. eGFP expression in erythroid cells peaks at E9.5 and then is rapidly silenced (>95%) and maintained at low levels into adulthood, demonstrating appropriate developmental regulation of the human ß-globin locus. In vitro knockdown of the epigenetic regulator DNA methyltransferase-1 in GG primary erythroid cells increases the proportion of eGFP(+) cells in culture from 41.9 to 74.1%. Furthermore, eGFP fluorescence is induced >3-fold after treatment of erythroid precursors with epigenetic drugs known to induce γ-globin expression, demonstrating the suitability of the Gγ-globin eGFP reporter for evaluation of γ-globin inducers. The GG mouse model is therefore a valuable model system for genetic and pharmacologic studies of the regulation of the ß-globin locus and for discovery of novel therapies for the ß-hemoglobinopathies.


Subject(s)
Erythropoiesis/physiology , Gene Expression Regulation , gamma-Globins/metabolism , Animals , DNA Modification Methylases/metabolism , Disease Models, Animal , Epigenesis, Genetic , Erythroid Cells/metabolism , Female , Fetal Hemoglobin/metabolism , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Promoter Regions, Genetic , RNA Interference , Time Factors , Transgenes , beta-Globins/metabolism
12.
FASEB J ; 28(4): 1610-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24371119

ABSTRACT

The clinical symptoms of hemoglobin disorders such as ß-thalassemia and sickle cell anemia are significantly ameliorated by the persistent expression of γ-globin after birth. This knowledge has driven the discovery of important regulators that silence γ-globin postnatally. Improved understanding of the γ- to ß-globin switching mechanism holds the key to devising targeted therapies for ß-hemoglobinopathies. To further investigate this mechanism, we used the murine erythroleukemic (MEL) cell line containing an intact 183-kb human ß-globin locus, in which the (G)γ- and ß-globin genes are replaced by DsRed and eGFP fluorescent reporters, respectively. Following RNA interference (RNAi)-mediated knockdown of two key transcriptional regulators, Myb and BCL11A, we observed a derepression of γ-globin, measured by DsRed fluorescence and qRT-PCR (P<0.001). Interestingly, double knockdown of Myb and DNA methyltransferase 1 (DNMT1) resulted in a robust induction of ε-globin, (up to 20% of total ß-like globin species) compared to single knockdowns (P<0.001). Conversely, double knockdowns of BCL11A and DNMT1 enhanced γ-globin expression (up to 90% of total ß-like globin species) compared to single knockdowns (P<0.001). Moreover, following RNAi treatment, expression of human ß-like globin genes mirrored the expression levels of their endogenous murine counterparts. These results demonstrate that Myb and BCL11A cooperate with DNMT1 to achieve developmental repression of embryonic and fetal ß-like globin genes in the adult erythroid environment.


Subject(s)
Carrier Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Fetal Hemoglobin/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myb/genetics , RNA Interference , Animals , Blotting, Western , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins , Erythropoiesis/genetics , Fetal Hemoglobin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transgenes/genetics , beta-Globins/genetics , beta-Globins/metabolism , epsilon-Globins/genetics , epsilon-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
13.
FASEB J ; 26(4): 1736-44, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267339

ABSTRACT

A greater understanding of the regulatory mechanisms that govern γ-globin expression in humans, especially the switching from γ- to ß-globin, which occurs after birth, would help to identify new therapeutic targets for patients with ß-hemoglobinopathy. To further elucidate the mechanisms involved in γ-globin expression, a novel fluorescent-based cellular reporter assay system was developed. Using homologous recombination, two reporter genes, DsRed and EGFP, were inserted into a 183-kb intact human ß-globin locus under the control of (G)γ- or (A)γ-globin promoter and ß-globin promoter, respectively. The modified constructs were stably transfected into adult murine erythroleukaemic (MEL) cells and human embryonic or fetal erythroleukemic (K562) cells, allowing for rapid and simultaneous analysis of fetal and adult globin gene expression according to their developmental stage-specific expression. To demonstrate the utility of this system, we performed RNA interference (RNAi)-mediated knockdown of BCL11A in the presence or absence of known fetal hemoglobin inducers and demonstrated functional derepression of a γ-globin-linked reporter in an adult erythroid environment. Our results demonstrate that the cellular assay system represents a promising approach to perform genetic and functional genomic studies to identify and evaluate key factors associated with γ-globin gene suppression.


Subject(s)
Gene Expression Regulation , Genes, Reporter , beta-Globins/genetics , gamma-Globins/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Fetal Hemoglobin/genetics , Fluorescent Dyes/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Repressor Proteins , Transgenes
14.
Novartis Found Symp ; 281: 38-43; discussion 44-53, 208-9, 2007.
Article in English | MEDLINE | ID: mdl-17534064

ABSTRACT

The lymphatic network functions to return fluid, cells and macromolecules to the circulation. Recent characterization of growth factors that control the growth and development of the lymphatics, and markers which specify lymphatic endothelial cells have enhanced our understanding of this system. Members of the VEGF family of factors are key regulators of these vessels with VEGF-C/VEGF-D and VEGFR-3 being the best validated signalling pathways in lymphangiogenesis. The study of these molecules in various pathologies has shown that they are important in the processes of cancer metastasis and in the formation of lymphoedema. Knowledge of these molecular pathways allows for the generation of modulators of these pathways which could form the basis of novel therapeutic approaches.


Subject(s)
Lymphangiogenesis/physiology , Neoplasms/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factors/metabolism , Humans , Lymphangiogenesis/genetics , Signal Transduction/genetics
15.
FASEB J ; 21(4): 1088-98, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17242158

ABSTRACT

Vascular endothelial growth factor (VEGF)-D is a secreted glycoprotein that induces angiogenesis and lymphangiogenesis. It consists of a central domain, containing binding sites for VEGF receptor-2 (VEGFR-2) and VEGFR-3, and N- and C-terminal propeptides. It is secreted from the cell as homodimers of the full-length form that can be proteolytically processed to remove the propeptides. It was recently shown, using adenoviral gene delivery, that fully processed VEGF-D induces angiogenesis in vivo, whereas full-length VEGF-D does not. To better understand these observations, we monitored the effect of VEGF-D processing on receptor binding using a full-length VEGF-D mutant that cannot be processed. This mutant binds VEGFR-2, the receptor signaling for angiogenesis, with approximately 17,000-fold lower affinity than mature VEGF-D, indicating the importance of processing for interaction with this receptor. Further, we show that members of the proprotein convertase (PC) family of proteases promote VEGF-D processing, which facilitates the VEGF-D/VEGFR-2 interaction. The PCs furin and PC5 promote cleavage of both propeptides, whereas PC7 promotes cleavage of the C-terminal propeptide only. The finding that PCs promote activation of VEGF-D and other proteins with roles in cancer such as matrix metalloproteinases, emphasizes the importance of these enzymes as potential regulators of tumor progression and metastasis.


Subject(s)
Carbamates/metabolism , Neovascularization, Pathologic , Oligopeptides/metabolism , Subtilisins/metabolism , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Glycoproteins/metabolism , HeLa Cells , Humans , Lymphatic System/pathology , Mice , Mice, Inbred BALB C , Mutation , Protein Binding , Vascular Endothelial Growth Factor D/chemistry
16.
Curr Cancer Drug Targets ; 5(8): 561-71, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16375662

ABSTRACT

The lymphatic vasculature is an important route of metastatic spread in cancer and recent studies have demonstrated that lymphangiogenesis (the growth of lymphatic vessels) associated with tumors promotes metastasis via the lymphatics. Therefore, the molecular mechanisms that drive lymphangiogenesis are attractive targets for development of novel therapeutics designed to restrict cancer metastasis. Such therapeutics would be of high priority as metastasis is the most lethal aspect of tumor biology. Research over the past seven years has identified protein growth factors and cell surface receptors that signal for lymphangiogenesis during embryonic development, in adult tissues and in cancer. Proteases that process and thereby activate lymphangiogenic growth factors have also been defined. Lymphangiogenic growth factors, the enzymes that activate them and the cell surface receptors signalling for growth of lymphatic vessels are prime targets for anti-lymphangiogenic drugs designed to restrict cancer metastasis. Agents targeting some of these proteins have already shown promise for blocking tumor lymphangiogenesis and lymphatic metastasis in animal models. This article focuses on current and emerging targets for blocking these processes that have been defined in recent studies of the molecular mechanisms controlling lymphangiogenesis. Strategies to block the actions of these proteins in cancer are also explored.


Subject(s)
Drug Delivery Systems/methods , Lymphangiogenesis/drug effects , Neoplasm Metastasis/prevention & control , Animals , Humans , Models, Biological , Receptors, Vascular Endothelial Growth Factor/drug effects , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/drug effects
18.
APMIS ; 112(7-8): 463-80, 2004.
Article in English | MEDLINE | ID: mdl-15563310

ABSTRACT

The vascular endothelial growth factor (VEGF) family of secreted glycoproteins are critical inducers of angiogenesis (growth of blood vessels) and lymphangiogenesis (growth of lymphatic vessels). These proteins are attractive therapeutic targets for blocking growth of blood vessels and lymphatics in tumors and thereby inhibiting the growth and spread of cancer -- in fact, the first VEGF inhibitor has recently entered the clinic for treatment of cancer. In addition, the VEGFs are being considered for stimulation of angiogenesis in the context of ischemic disease and lymphangiogenesis for treatment of lymphedema. These therapeutic possibilities have focused great interest on the molecular regulation of VEGF family members. Much has been learned in the past five years about the mechanisms controlling the action of the VEGFs, including the importance of hypoxia, proteolysis, transcription factors and RNA splicing. An understanding of these mechanisms offers broader opportunities to manipulate expression and activity of the VEGFs for treatment of disease.


Subject(s)
Lymphangiogenesis , Neovascularization, Pathologic/etiology , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Communication , Cell Transformation, Neoplastic/genetics , Embryonic Development , Humans , Membrane Proteins , Proteins/physiology , Proto-Oncogene Proteins c-fos/physiology
19.
J Exp Med ; 198(6): 863-8, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12963694

ABSTRACT

Vascular endothelial growth factor (VEGF) C and VEGF-D stimulate lymphangiogenesis and angiogenesis in tissues and tumors by activating the endothelial cell surface receptor tyrosine kinases VEGF receptor (VEGFR) 2 and VEGFR-3. These growth factors are secreted as full-length inactive forms consisting of NH2- and COOH-terminal propeptides and a central VEGF homology domain (VHD) containing receptor binding sites. Proteolytic cleavage removes the propeptides to generate mature forms, consisting of dimers of the VEGF homology domain, that bind receptors with much greater affinity than the full-length forms. Therefore, proteolytic processing activates VEGF-C and VEGF-D, although the proteases involved were unknown. Here, we report that the serine protease plasmin cleaved both propeptides from the VEGF homology domain of human VEGF-D and thereby generated a mature form exhibiting greatly enhanced binding and cross-linking of VEGFR-2 and VEGFR-3 in comparison to full-length material. Plasmin also activated VEGF-C. As lymphangiogenic growth factors promote the metastatic spread of cancer via the lymphatics, the proteolytic activation of these molecules represents a potential target for antimetastatic agents. Identification of an enzyme that activates the lymphangiogenic growth factors will facilitate development of inhibitors of metastasis.


Subject(s)
Endothelial Growth Factors/metabolism , Fibrinolysin/metabolism , Lymphatic System/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Endothelial Growth Factors/genetics , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor D , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...