Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 22(14)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34299378

ABSTRACT

The importance of lymphatic vessels in a myriad of human diseases is rapidly gaining recognition; lymphatic vessel dysfunction is a feature of disorders including congenital lymphatic anomalies, primary lymphoedema and obesity, while improved lymphatic vessel function increases the efficacy of immunotherapy for cancer and neurological disease and promotes cardiac repair following myocardial infarction. Understanding how the growth and function of lymphatic vessels is precisely regulated therefore stands to inform the development of novel therapeutics applicable to a wide range of human diseases. Lymphatic vascular development is initiated during embryogenesis following establishment of the major blood vessels and the onset of blood flow. Lymphatic endothelial progenitor cells arise from a combination of venous and non-venous sources to generate the initial lymphatic vascular structures in the vertebrate embryo, which are then further ramified and remodelled to elaborate an extensive lymphatic vascular network. Signalling mediated via vascular endothelial growth factor (VEGF) family members and vascular endothelial growth factor receptor (VEGFR) tyrosine kinases is crucial for development of both the blood and lymphatic vascular networks, though distinct components are utilised to different degrees in each vascular compartment. Although much is known about the regulation of VEGFA/VEGFR2 signalling in the blood vasculature, less is understood regarding the mechanisms by which VEGFC/VEGFD/VEGFR3 signalling is regulated during lymphatic vascular development. This review will focus on recent advances in our understanding of the cellular and molecular mechanisms regulating VEGFA-, VEGFC- and VEGFD-mediated signalling via VEGFRs which are important for driving the construction of lymphatic vessels during development and disease.


Subject(s)
Lymphatic Vessels/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/physiology , Animals , Endothelial Cells/metabolism , Humans , Lymphangiogenesis/physiology
2.
BMC Dev Biol ; 17(1): 5, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28407732

ABSTRACT

BACKGROUND: Cranial neural crest cells (NCCs) are a unique embryonic cell type which give rise to a diverse array of derivatives extending from neurons and glia through to bone and cartilage. Depending on their point of origin along the antero-posterior axis cranial NCCs are rapidly sorted into distinct migratory streams that give rise to axial specific structures. These migratory streams mirror the underlying segmentation of the brain with NCCs exiting the diencephalon and midbrain following distinct paths compared to those exiting the hindbrain rhombomeres (r). The genetic landscape of cranial NCCs arising at different axial levels remains unknown. RESULTS: Here we have used RNA sequencing to uncover the transcriptional profiles of mouse cranial NCCs arising at different axial levels. Whole transcriptome analysis identified over 120 transcripts differentially expressed between NCCs arising anterior to r3 (referred to as r1-r2 migratory stream for simplicity) and the r4 migratory stream. Eight of the genes differentially expressed between these populations were validated by RT-PCR with 2 being further validated by in situ hybridisation. We also explored the expression of the Neuropilins (Nrp1 and Nrp2) and their co-receptors and show that the A-type Plexins are differentially expressed in different cranial NCC streams. CONCLUSIONS: Our analyses identify a large number of genes differentially regulated between cranial NCCs arising at different axial levels. This data provides a comprehensive description of the genetic landscape driving diversity of distinct cranial NCC streams and provides novel insight into the regulatory networks controlling the formation of specific skeletal elements and the mechanisms promoting migration along different paths.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Neural Crest/cytology , Neural Crest/growth & development , Sequence Analysis, RNA/methods , Animals , Cell Movement , Diencephalon/cytology , Diencephalon/growth & development , Gene Expression Regulation, Developmental , Mesencephalon/cytology , Mesencephalon/growth & development , Mice , Nerve Tissue Proteins/genetics , Neuropilin-1/genetics , Neuropilin-2/genetics , Rhombencephalon/cytology , Rhombencephalon/growth & development
3.
Dev Dyn ; 244(3): 323-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25399804

ABSTRACT

Lymphatic vessels are an integral component of the cardiovascular system, serving important roles in fluid homeostasis, lipid absorption, and immune cell trafficking. Defining the mechanisms by which the lymphatic vasculature is constructed and remodeled into a functional vascular network not only provides answers to fascinating biological questions, but is fundamental to understanding how lymphatic vessel growth and development goes awry in human pathologies. While long recognized as dysfunctional in lymphedema and exploited as a route of tumor metastasis, recent work has highlighted important roles for lymphatic vessels in modulating immune responses, regulating salt-sensitive hypertension and important for lung inflation at birth. Substantial progress in our understanding of the signaling pathways important for development and morphogenesis of the lymphatic vasculature has been made in recent years. Here, we review advances in our knowledge of the best characterized of these signaling pathways, that involving the vascular endothelial growth factor (VEGF) family members VEGF-C and VEGF-D, together with their receptors VEGFR2 and VEGFR3. Recent work has defined multiple levels at which signal transduction by means of this key axis is regulated; these include control of ligand processing and bioavailability, modulation of receptor activation by interacting proteins, and regulation of receptor endocytosis and trafficking.


Subject(s)
Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Signal Transduction/physiology , Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Humans , Lymphatic Vessels/cytology , Stem Cells/cytology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism
4.
Dev Biol ; 386(1): 25-33, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24361262

ABSTRACT

During embryogenesis, lymphatic endothelial progenitor cells first arise from a subset of blood vascular endothelial cells in the dorsolateral aspects of the cardinal veins. The molecular cues responsible for defining the regionalisation of such a discrete pool of progenitors remain uncharacterised. Here we identify a novel function for CYP26B1, an enzyme known to play a role in tissue morphogenesis by fine-tuning retinoic acid (RA) concentration, in regulating lymphangiogenesis. Cyp26b1-null mice, in which RA levels are elevated, exhibited an increased number of lymphatic endothelial progenitor cells in the cardinal veins, together with hyperplastic, blood filled lymph sacs and hyperplastic dermal lymphatic vessels. Conversely, mice over-expressing Cyp26b1 had hypoplastic lymph sacs and lymphatic vessels. Our data suggest that RA clearance by CYP26B1 in the vicinity of lymphatic endothelial progenitor cells is important for determining the position and size of the progenitor pool specified. Our studies identify a genetic pathway that underpins the architecture of the developing lymphatics and define CYP26B1 as a novel modulator of lymphatic vascular patterning.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Developmental , Lymphangiogenesis , Lymphatic System/embryology , Lymphatic Vessels/metabolism , Retinoids/metabolism , Animals , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Endothelial Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Phenotype , Retinoic Acid 4-Hydroxylase , Signal Transduction , Transgenes , Tretinoin/metabolism
5.
Dev Biol ; 383(2): 186-200, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24080509

ABSTRACT

The integration of multiple morphogenic signalling pathways and transcription factor networks is essential to mediate neural crest (NC) cell induction, delamination, survival, stem-cell properties, fate choice and differentiation. Although the transcriptional control of NC development is well documented in mammals, the role of post-transcriptional modifications, and in particular ubiquitination, has not been explored. Here we report an essential role for the ubiquitin ligase Nedd4 in cranial NC cell development. Our analysis of Nedd4(-/-) embryos identified profound deficiency of cranial NC cells in the absence of structural defects in the neural tube. Nedd4 is expressed in migrating cranial NC cells and was found to positively regulate expression of the NC transcription factors Sox9, Sox10 and FoxD3. We found that in the absence of these factors, a subset of cranial NC cells undergo apoptosis. In accordance with a lack of cranial NC cells, Nedd4(-/-) embryos have deficiency of the trigeminal ganglia, NC derived bone and malformation of the craniofacial skeleton. Our analyses therefore uncover an essential role for Nedd4 in a subset of cranial NC cells and highlight E3 ubiquitin ligases as a likely point of convergence for multiple NC signalling pathways and transcription factor networks.


Subject(s)
Brain/cytology , Brain/embryology , Endosomal Sorting Complexes Required for Transport/metabolism , Face/embryology , Neural Crest/cytology , Stem Cells/cytology , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Body Patterning , Cell Proliferation , Cell Survival , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endosomal Sorting Complexes Required for Transport/deficiency , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Nedd4 Ubiquitin Protein Ligases , Phenotype , Rhombencephalon/cytology , Rhombencephalon/embryology , Stem Cells/metabolism , Transcription Factors/metabolism , Trigeminal Ganglion/cytology , Trigeminal Ganglion/embryology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
6.
Blood ; 119(5): 1283-91, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22147895

ABSTRACT

Recent work has established that heterozygous germline GATA2 mutations predispose carriers to familial myelodysplastic syndrome (MDS)/acute myeloid leukemia (AML), "MonoMAC" syndrome, and DCML deficiency. Here, we describe a previously unreported MDS family carrying a missense GATA2 mutation (p.Thr354Met), one patient with MDS/AML carrying a frameshift GATA2 mutation (p.Leu332Thrfs*53), another with MDS harboring a GATA2 splice site mutation, and 3 patients exhibiting MDS or MDS/AML who have large deletions encompassing the GATA2 locus. Intriguingly, 2 MDS/AML or "MonoMAC" syndrome patients with GATA2 deletions and one with a frameshift mutation also have primary lymphedema. Primary lymphedema occurs as a result of aberrations in the development and/or function of lymphatic vessels, spurring us to investigate whether GATA2 plays a role in the lymphatic vasculature. We demonstrate here that GATA2 protein is present at high levels in lymphatic vessel valves and that GATA2 controls the expression of genes important for programming lymphatic valve development. Our data expand the phenotypes associated with germline GATA2 mutations to include predisposition to primary lymphedema and suggest that complete haploinsufficiency or loss of function of GATA2, rather than missense mutations, is the key predisposing factor for lymphedema onset. Moreover, we reveal a crucial role for GATA2 in lymphatic vascular development.


Subject(s)
GATA2 Transcription Factor/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Lymphatic Vessels/metabolism , Lymphedema/congenital , Myelodysplastic Syndromes/genetics , Adolescent , Adult , Animals , Cells, Cultured , Child , Female , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/physiology , Germ-Line Mutation/physiology , Humans , Infant, Newborn , Lymphangiogenesis/genetics , Lymphedema/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/pathology , Syndrome , Young Adult
7.
Dev Biol ; 364(2): 89-98, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22230615

ABSTRACT

During lymphangiogenesis in the mammalian embryo, a subset of vascular endothelial cells in the cardinal veins is reprogrammed to adopt a lymphatic endothelial fate. The prevailing model of lymphangiogenesis contends that these lymphatic precursor cells migrate away from the cardinal veins and reassemble peripherally as lymph sacs from which a lymphatic vasculature is generated. However, this model fails to account for a number of observations that, as a result, have remained anecdotal. Here, we use optical projection tomography, confocal microscopy and in vivo live imaging to uncover three key stages of lymphatic vascular morphogenesis in the mouse embryo at high resolution. First, we define territories or "pre-lymphatic clusters" of Prox1-positive lymphatic endothelial progenitor cells along the antero-posterior axis of the cardinal veins. Second, these pre-lymphatic clusters undergo progressive extrusion ("ballooning") to generate primitive lymph sacs. Third, lymphatic vessels emerge by a combination of mechanisms including sprouting from the lymph sacs and direct delamination of streams of cells from the cardinal veins. Our data support a new model for lymphatic vascular patterning and morphogenesis, as a basis for identifying the molecular cues governing these processes.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels/embryology , Veins/embryology , Animals , Homeodomain Proteins/analysis , Mice , Tumor Suppressor Proteins/analysis
8.
Science ; 381(6659): 799-804, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590348

ABSTRACT

Piezo channels are critical cellular sensors of mechanical forces. Despite their large size, ubiquitous expression, and irreplaceable roles in an ever-growing list of physiological processes, few Piezo channel-binding proteins have emerged. In this work, we found that MyoD (myoblast determination)-family inhibitor proteins (MDFIC and MDFI) are PIEZO1/2 interacting partners. These transcriptional regulators bind to PIEZO1/2 channels, regulating channel inactivation. Using single-particle cryogenic electron microscopy, we mapped the interaction site in MDFIC to a lipidated, C-terminal helix that inserts laterally into the PIEZO1 pore module. These Piezo-interacting proteins fit all the criteria for auxiliary subunits, contribute to explaining the vastly different gating kinetics of endogenous Piezo channels observed in many cell types, and elucidate mechanisms potentially involved in human lymphatic vascular disease.


Subject(s)
Ion Channels , Myogenic Regulatory Factors , Humans , Cryoelectron Microscopy , HEK293 Cells , Ion Channel Gating , Ion Channels/chemistry , Ion Channels/genetics , Ion Channels/metabolism , Kinetics , Lymphatic Diseases/genetics , Mutation , Myogenic Regulatory Factors/chemistry , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Protein Domains , Myoblasts/metabolism , Animals , Mice
9.
Nat Commun ; 13(1): 2018, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440627

ABSTRACT

The ubiquitin ligase NEDD4 promotes neural crest cell (NCC) survival and stem-cell like properties to regulate craniofacial and peripheral nervous system development. However, how ubiquitination and NEDD4 control NCC development remains unknown. Here we combine quantitative analysis of the proteome, transcriptome and ubiquitinome to identify key developmental signalling pathways that are regulated by NEDD4. We report 276 NEDD4 targets in NCCs and show that loss of NEDD4 leads to a pronounced global reduction in specific ubiquitin lysine linkages. We further show that NEDD4 contributes to the regulation of the NCC actin cytoskeleton by controlling ubiquitination and turnover of Profilin 1 to modulate filamentous actin polymerization. Taken together, our data provide insights into how NEDD4-mediated ubiquitination coordinates key regulatory processes during NCC development.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Neural Crest , Actins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Neural Crest/metabolism , Profilins/genetics , Profilins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Sci Transl Med ; 14(634): eabm4869, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35235341

ABSTRACT

Central conducting lymphatic anomaly (CCLA), characterized by the dysfunction of core collecting lymphatic vessels including the thoracic duct and cisterna chyli, and presenting as chylothorax, pleural effusions, chylous ascites, and lymphedema, is a severe disorder often resulting in fetal or perinatal demise. Although pathogenic variants in RAS/mitogen activated protein kinase (MAPK) signaling pathway components have been documented in some patients with CCLA, the genetic etiology of the disorder remains uncharacterized in most cases. Here, we identified biallelic pathogenic variants in MDFIC, encoding the MyoD family inhibitor domain containing protein, in seven individuals with CCLA from six independent families. Clinical manifestations of affected fetuses and children included nonimmune hydrops fetalis (NIHF), pleural and pericardial effusions, and lymphedema. Generation of a mouse model of human MDFIC truncation variants revealed that homozygous mutant mice died perinatally exhibiting chylothorax. The lymphatic vasculature of homozygous Mdfic mutant mice was profoundly mispatterned and exhibited major defects in lymphatic vessel valve development. Mechanistically, we determined that MDFIC controls collective cell migration, an important early event during the formation of lymphatic vessel valves, by regulating integrin ß1 activation and the interaction between lymphatic endothelial cells and their surrounding extracellular matrix. Our work identifies MDFIC variants underlying human lymphatic disease and reveals a crucial, previously unrecognized role for MDFIC in the lymphatic vasculature. Ultimately, understanding the genetic and mechanistic basis of CCLA will facilitate the development and implementation of new therapeutic approaches to effectively treat this complex disease.


Subject(s)
Chylothorax , Lymphatic Vessels , Lymphedema , Myogenic Regulatory Factors , Animals , Chylothorax/genetics , Chylothorax/metabolism , Endothelial Cells , Female , Humans , Hydrops Fetalis/genetics , Hydrops Fetalis/metabolism , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/metabolism , Mice , Myogenic Regulatory Factors/genetics , Pregnancy
11.
J Clin Invest ; 130(6): 3315-3328, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32182215

ABSTRACT

The atypical cadherin FAT4 has established roles in the regulation of planar cell polarity and Hippo pathway signaling that are cell context dependent. The recent identification of FAT4 mutations in Hennekam syndrome, features of which include lymphedema, lymphangiectasia, and mental retardation, uncovered an important role for FAT4 in the lymphatic vasculature. Hennekam syndrome is also caused by mutations in collagen and calcium binding EGF domains 1 (CCBE1) and ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3), encoding a matrix protein and protease, respectively, that regulate activity of the key prolymphangiogenic VEGF-C/VEGFR3 signaling axis by facilitating the proteolytic cleavage and activation of VEGF-C. The fact that FAT4, CCBE1, and ADAMTS3 mutations underlie Hennekam syndrome suggested that all 3 genes might function in a common pathway. We identified FAT4 as a target gene of GATA-binding protein 2 (GATA2), a key transcriptional regulator of lymphatic vascular development and, in particular, lymphatic vessel valve development. Here, we demonstrate that FAT4 functions in a lymphatic endothelial cell-autonomous manner to control cell polarity in response to flow and is required for lymphatic vessel morphogenesis throughout development. Our data reveal a crucial role for FAT4 in lymphangiogenesis and shed light on the mechanistic basis by which FAT4 mutations underlie a human lymphedema syndrome.


Subject(s)
Cadherins/metabolism , Cell Polarity , Endothelial Cells/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Animals , Cadherins/genetics , Endothelial Cells/pathology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Humans , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/metabolism , Lymphedema/pathology , Mice , Mice, Transgenic , Syndrome
12.
Dev Cell ; 49(2): 279-292.e5, 2019 04 22.
Article in English | MEDLINE | ID: mdl-31014480

ABSTRACT

The correct assignment of cell fate within fields of multipotent progenitors is essential for accurate tissue diversification. The first lymphatic vessels arise from pre-existing veins after venous endothelial cells become specified as lymphatic progenitors. Prox1 specifies lymphatic fate and labels these progenitors; however, the mechanisms restricting Prox1 expression and limiting the progenitor pool remain unknown. We identified a zebrafish mutant that displayed premature, expanded, and prolonged lymphatic specification. The gene responsible encodes the regulator of alternative splicing, Nova2. In zebrafish and human endothelial cells, Nova2 selectively regulates pre-mRNA splicing for components of signaling pathways and phosphoproteins. Nova2-deficient endothelial cells display increased Mapk/Erk signaling, and Prox1 expression is dynamically controlled by Erk signaling. We identify a mechanism whereby Nova2-regulated splicing constrains Erk signaling, thus limiting lymphatic progenitor cell specification. This identifies the capacity of a factor that tunes mRNA splicing to control assignment of cell fate during vascular differentiation.


Subject(s)
Lymphatic Vessels/metabolism , MAP Kinase Signaling System , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Cell Differentiation , Cell Lineage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Lymphangiogenesis , Lymphatic Vessels/cytology , Male , Neuro-Oncological Ventral Antigen , Tumor Suppressor Proteins/metabolism , Veins/cytology , Veins/metabolism , Zebrafish
13.
Ophthalmology ; 115(11): 1989-97, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18554721

ABSTRACT

OBJECTIVE: To determine, using objective measures, the outcome of ex vivo cultured limbal epithelial stem cell (LESC) transplantation performed in compliance with good manufacturing practice using a novel culture system without 3T3 feeder cells. DESIGN: Prospective, noncomparative, interventional case series. PARTICIPANTS: Ten eyes of 10 patients with profound LESC deficiency arising from chemical injury (4 eyes), aniridia (3 eyes), ectodermal dysplasia (1 eye), Reiger's anomaly with Pax6 haploinsufficiency (1 eye), and unknown cause (1 eye). METHODS: Allogeneic (7 eyes) or autologous (3 eyes) corneal LESCs were cultured on human amniotic membrane. Tissue was transplanted to the recipient eye after superficial keratectomy. Impression cytology and confocal microscopy were performed 6 months after surgery with clinical follow-up to 13 months. Success was defined as an improvement in the defined clinical parameters of LESC deficiency, an improvement in visual acuity, the restoration of a more normal corneal phenotype on impression cytology, and the appearance of a regular hexagonal basal layer of cells on corneal confocal microscopy. MAIN OUTCOME MEASURES: Clinical parameters of LESC deficiency (loss of epithelial transparency, superficial corneal vascularization, epithelial irregularity, and epithelial breakdown), visual acuity, impression cytology and cytokeratin expression profiles, and in vivo confocal corneal confocal microscopy. RESULTS: The success rate using this technique was 60% (autografts 33%, allografts 71%). All patients with a successful outcome experienced an improvement in visual acuity of >/=2 lines Snellen acuity. Preoperatively, CK3+ and CK19+ cells accounted for 12+/-2.4% (mean +/- standard error of the mean) and 80+/-2.15% of cells, respectively, whereas postoperatively these accounted for 69+/-6.43% (P<0.0001) and 30+/-6.34% (P<0.0001) of cells, respectively. Goblet cells accounted for 8+/-1.19% of cells preoperatively and 1+/-0.35% of cells postoperatively (P<0.0001). CONCLUSIONS: These data demonstrate that it is possible to culture LESCs ex vivo in compliance with good manufacturing practice regulations. A set of objective outcome measures that confirm the efficiency of this technique in treating LESC deficiency is described. The widespread use of such standardized and objective outcome measures would facilitate a comparison between the different culture methods in use.


Subject(s)
Corneal Diseases/surgery , Epithelium, Corneal/cytology , Limbus Corneae/cytology , Stem Cell Transplantation , Stem Cells/cytology , Adult , Aged , Cell Count , Cell Transplantation/methods , Cells, Cultured , Corneal Diseases/pathology , Female , Humans , Immunosuppressive Agents/administration & dosage , Male , Microscopy, Confocal , Middle Aged , Prospective Studies , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome , Visual Acuity
14.
Surv Ophthalmol ; 52(5): 483-502, 2007.
Article in English | MEDLINE | ID: mdl-17719371

ABSTRACT

Ex vivo cultured limbal epithelial stem cells have been used successfully to treat corneal limbal stem cell deficiency. We identified 17 reports of the application of this novel cell-based therapy in humans. In addition we identified four reports of the use of culture oral mucosal epithelial cells to treat limbal stem cell deficiency. We examined these reports to discern the success rate, complication rate, visual outcome, whether there is an optimal technique and which patients are the most likely to benefit. We also discuss the different culture methods employed and the regulations governing cell banks that are providing this service. We found that the techniques used to cultivate and transplant cells varied, but that no individual method was clearly superior. The reported success rate is similar across all studies for both allografts and autografts. The clinical indications for this treatment are not clearly defined as indicated by the variety of disorders treated. Follow-up is limited and the long-term success rate is yet to be established. Nonetheless, we conclude that there is sufficient evidence to support the continued use and refinement of this procedure as a treatment for corneal stem cell deficiency.


Subject(s)
Corneal Diseases/surgery , Epithelium, Corneal/cytology , Stem Cell Transplantation/methods , Cells, Cultured , Epithelial Cells/transplantation , Humans
15.
Cancer Res ; 62(16): 4599-604, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12183414

ABSTRACT

Numerous cytogenetic and molecular studies of breast cancer have identified frequent loss of heterozygosity (LOH) of the long arm of human chromosome 16. On the basis of these data, the likely locations of breast cancer tumor suppressor genes are bands 16q22.1 and 16q24.3. We have mapped the CBFA2T3 (MTG16) gene, previously cloned as a fusion partner of the AML1 protein from a rare (16;21) leukemia translocation, to the 16q24.3 breast cancer LOH region. The expression of CBFA2T3 was significantly reduced in a number of breast cancer cell lines and in primary breast tumors, including early ductal carcinomas in situ, when compared with nontransformed breast epithelial cell lines and normal breast tissue. Reintroduction of CBFA2T3 into different breast tumor derived cell lines with decreased expression of this gene reduced colony growth on plastic and in soft agar. CBFA2T3 was shown to function as a transcriptional repressor when tethered to the GAL4 DNA-binding domain in a reporter gene assay and, therefore, has the potential to be a transcriptional repressor in normal breast epithelial cells. Taken together, these findings suggest that CBFA2T3 is a likely candidate for the breast cancer tumor suppressor gene that is the target for the frequent 16q24 LOH in breast neoplasms.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 16/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity , Phosphoproteins , Proteins/genetics , Tumor Suppressor Proteins , Cell Division/genetics , Gene Expression Regulation, Neoplastic , Humans , Protein Biosynthesis , Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Repressor Proteins , Tumor Cells, Cultured
16.
J Clin Invest ; 125(8): 2979-94, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26214525

ABSTRACT

Heterozygous germline mutations in the zinc finger transcription factor GATA2 have recently been shown to underlie a range of clinical phenotypes, including Emberger syndrome, a disorder characterized by lymphedema and predisposition to myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). Despite well-defined roles in hematopoiesis, the functions of GATA2 in the lymphatic vasculature and the mechanisms by which GATA2 mutations result in lymphedema have not been characterized. Here, we have provided a molecular explanation for lymphedema predisposition in a subset of patients with germline GATA2 mutations. Specifically, we demonstrated that Emberger-associated GATA2 missense mutations result in complete loss of GATA2 function, with respect to the capacity to regulate the transcription of genes that are important for lymphatic vessel valve development. We identified a putative enhancer element upstream of the key lymphatic transcriptional regulator PROX1 that is bound by GATA2, and the transcription factors FOXC2 and NFATC1. Emberger GATA2 missense mutants had a profoundly reduced capacity to bind this element. Conditional Gata2 deletion in mice revealed that GATA2 is required for both development and maintenance of lymphovenous and lymphatic vessel valves. Together, our data unveil essential roles for GATA2 in the lymphatic vasculature and explain why a select catalogue of human GATA2 mutations results in lymphedema.


Subject(s)
GATA2 Transcription Factor/metabolism , Lymphatic Vessels/embryology , Lymphedema/embryology , Mutation , Animals , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/genetics , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/pathology , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Stem Cells Transl Med ; 3(2): 265-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24443006

ABSTRACT

Limbal stem cell deficiency (LSCD) is an eye disorder in which the stem cells responsible for forming the surface skin of the cornea are destroyed by disease. This results in pain, loss of vision, and a cosmetically unpleasant appearance. Many new treatments, including stem cell therapies, are emerging for the treatment of this condition, but assessment of these new technologies is severely hampered by the lack of biomarkers for this disease or validated tools for assessing its severity. The aims of this study were to design and test the reliability of a tool for grading LSCD, to define a set of core outcome measures for use in evaluating treatments for this condition, and to demonstrate their utility. This was achieved by using our defined outcome set (which included the Clinical Outcome Assessment in Surgical Trials of Limbal stem cell deficiency [COASTL] tool) to evaluate the 3-year outcomes for allogeneic ex vivo cultivated limbal epithelial transplantation (allo-CLET) in patients who had bilateral total LSCD secondary to aniridia or Stevens-Johnson syndrome. The results demonstrate that our new grading tool for LSCD, the COASTL tool, is reliable and repeatable, and that improvements in the biomarkers used in this tool correlate positively with improvements in visual acuity. The COASTL tool showed that following allo-CLET there was a decrease in LSCD severity and an increase in visual acuity up to 12 months post-treatment, but thereafter LSCD severity and visual acuity progressively deteriorated.


Subject(s)
Aniridia/surgery , Epithelium, Corneal/pathology , Limbus Corneae/pathology , Postoperative Complications/pathology , Severity of Illness Index , Stevens-Johnson Syndrome/pathology , Allografts , Aniridia/pathology , Biomarkers/metabolism , Cells, Cultured , Corneal Opacity/pathology , Corneal Opacity/surgery , Corneal Transplantation/methods , Databases, Factual , Epithelium, Corneal/surgery , Follow-Up Studies , Humans , Limbus Corneae/surgery , Reproducibility of Results , Stem Cell Transplantation/methods , Stevens-Johnson Syndrome/surgery , Treatment Outcome
18.
PLoS One ; 7(7): e40497, 2012.
Article in English | MEDLINE | ID: mdl-22792354

ABSTRACT

Despite the importance of blood vessels and lymphatic vessels during development and disease, the signalling pathways underpinning vessel construction remain poorly characterised. Primary mouse endothelial cells have traditionally proven difficult to culture and as a consequence, few assays have been developed to dissect gene function and signal transduction pathways in these cells ex vivo. Having established methodology for the purification, short-term culture and transfection of primary blood (BEC) and lymphatic (LEC) vascular endothelial cells isolated from embryonic mouse skin, we sought to optimise robust assays able to measure embryonic LEC proliferation, migration and three-dimensional tube forming ability in vitro. In the course of developing these assays using the pro-lymphangiogenic growth factors FGF2 and VEGF-C, we identified previously unrecognised roles for FGFR1 signalling in lymphangiogenesis. The small molecule FGF receptor tyrosine kinase inhibitor SU5402, but not inhibitors of VEGFR-2 (SU5416) or VEGFR-3 (MAZ51), inhibited FGF2 mediated LEC proliferation, demonstrating that FGF2 promotes proliferation directly via FGF receptors and independently of VEGF receptors in primary embryonic LEC. Further investigation revealed that FGFR1 was by far the predominant FGF receptor expressed by primary embryonic LEC and correspondingly, siRNA-mediated FGFR1 knockdown abrogated FGF2 mediated LEC proliferation. While FGF2 potently promoted LEC proliferation and migration, three dimensional tube formation assays revealed that VEGF-C primarily promoted LEC sprouting and elongation, illustrating that FGF2 and VEGF-C play distinct, cooperative roles in lymphatic vascular morphogenesis. These assays therefore provide useful tools able to dissect gene function in cellular events important for lymphangiogenesis and implicate FGFR1 as a key player in developmental lymphangiogenesis in vivo.


Subject(s)
Endothelial Cells/metabolism , Lymphangiogenesis/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Separation , Endothelial Cells/cytology , Female , Fibroblast Growth Factor 2/pharmacology , Lymphangiogenesis/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , Vascular Endothelial Growth Factor C/pharmacology
19.
Invest Ophthalmol Vis Sci ; 53(13): 8076-85, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23139271

ABSTRACT

PURPOSE: To investigate signal transduction pathways for connective tissue growth factor (CTGF) in human corneal fibroblasts (HCF). METHODS: Expression of 75 kinases in cultures of serum-starved (HCF) were investigated using protein kinase screens, and changes in levels of phosphorylation of 31 different phosphoproteins were determined at 0, 5, and 15 minutes after treatment with CTGF. Levels of phosphorylation of three signal transducing phosphoproteins (extracellular regulated kinase 1 [ERK1], extracellular regulated kinase 2 [ERK2] [MAPKs], and signal transducer and activator of transcription 3 [STAT3]) were measured at nine time points after exposure to CTGF using Western immunoblots. Inhibition of Ras, MEK1/2 (MAPKK), and ERK1/2, on CTGF-stimulated fibroblast proliferation and collagen gel contraction was assessed using selective inhibitors farnesylthiosalicylic acid, PD-98059, and SB203580, respectively. RESULTS: Thirty two of the 75 kinases (43%) evaluated by the kinase screen were detected in extracts of quiescent HCF, suggesting these kinases are available to respond acutely to CTGF exposure. Addition of CTGF increased levels of phosphorylation of five phosphoproteins (ERK1 and 2, MEK1/2 [MAPKK], STAT3, and SAPK/JNK), and decreased levels of phosphorylation of 14 phosphoproteins (including protein kinases B and C) after 5 and 15 minutes. Further analysis of ERK1 and 2 and STAT3 phosphorylation showed rapid increases within 1 minute of CTGF exposure that peaked between 5 and 10 minutes then returned to pretreatment levels by 30 minutes. Treatment of HCF with selective inhibitors of Ras, MEK 1/2, and ERK1/2 individually blocked both CTGF induced cell proliferation, and collagen gel contraction. CONCLUSIONS: Results from protein kinase screens and selective kinase inhibitors demonstrate Ras/MEK/ERK/STAT3 pathway is required for CTGF signaling in HCF.


Subject(s)
Connective Tissue Growth Factor/pharmacology , Corneal Keratocytes/drug effects , Protein Kinases/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Corneal Keratocytes/enzymology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , ras Proteins/metabolism
20.
Biomaterials ; 30(6): 1056-65, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19019426

ABSTRACT

Human amniotic membrane (HAM) is employed as a substrate for the ex-vivo expansion of limbal epithelial cells (LECs) used to treat corneal epithelial stem cell deficiency in humans. The optimal method of HAM preparation for this purpose is unknown. This study evaluated the ability of different preparations of stored HAM to serve as substrates for LEC expansion ex-vivo. The effect of removing the amniotic epithelial cells (decellularisation) from HAM prior to seeding of LECs, the effect of glycerol cryopreservation and the effect of peracetic acid (PAA) sterilization and antibiotic disinfection were evaluated using different HAM test groups. Human LECs were cultured on each preparation and the following outcomes were assessed: confluence of growth, cell density, cell morphology and expression of the putative LESC markers deltaN-p63alpha and ABCG2. Removing amniotic epithelial cells prior to seeding of LECs resulted in a higher percentage of confluence but a lower cell density than intact HAM suggesting that decellularisation does not increase proliferation, but rather that it facilitates migration of LECs resulting in larger cells. Decellularisation did not affect the percentage of cells expressing the putative LESC markers deltaN-p63alpha (< or =4% in both intact and acellular groups) and ABCG2 (< or =3% in both intact and acellular groups). Glycerol cryopreservation of HAM resulted in poor morphology and a low proportion of cells expressing deltaN-p63alpha (< or =6%) and ABCG2 (< or =8%). HAM frozen at -80 degrees C in Hank's Balanced Salt Solution (HBSS) was superior, demonstrating excellent morphology of cultured LECs and high levels of deltaN-p63alpha (< or =68%) and ABCG2 (< or =62%) expression (p<0.001). The use of PAA or antibiotics to decontaminate HAM does not appear to affect this function. The variables affecting the ability of HAM to serve as a substrate for LEC expansion ex-vivo are poorly understood. The use of glycerol as a cryoprotectant impairs this ability whereas simple frozen HAM appears to work extremely well for this purpose.


Subject(s)
Amnion/metabolism , Epithelial Cells/cytology , Limbus Corneae/cytology , Materials Testing/methods , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Anti-Bacterial Agents/pharmacology , Cell Count , Cell Proliferation/drug effects , Cell Shape/drug effects , Cryopreservation , Epithelial Cells/drug effects , Glycerol , Humans , Neoplasm Proteins/metabolism , Time Factors , Trans-Activators/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL