Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Commun ; 15(1): 6613, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098861

ABSTRACT

Tumour-host immune interactions lead to complex changes in the tumour microenvironment (TME), impacting progression, metastasis and response to therapy. While it is clear that cancer cells can have the capacity to alter immune landscapes, our understanding of this process is incomplete. Herein we show that endocytic trafficking at the plasma membrane, mediated by the small GTPase ARF6, enables melanoma cells to impose an immunosuppressive TME that accelerates tumour development. This ARF6-dependent TME is vulnerable to immune checkpoint blockade therapy (ICB) but in murine melanoma, loss of Arf6 causes resistance to ICB. Likewise, downregulation of ARF6 in patient tumours correlates with inferior overall survival after ICB. Mechanistically, these phenotypes are at least partially explained by ARF6-dependent recycling, which controls plasma membrane density of the interferon-gamma receptor. Collectively, our findings reveal the importance of endomembrane trafficking in outfitting tumour cells with the ability to shape their immune microenvironment and respond to immunotherapy.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Cell Membrane , Immune Checkpoint Inhibitors , Melanoma , Tumor Microenvironment , Tumor Microenvironment/immunology , Animals , Humans , Mice , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Melanoma/immunology , Cell Line, Tumor , Cell Membrane/metabolism , Interferon gamma Receptor , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Protein Transport , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/genetics , Mice, Inbred C57BL , Female
2.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873189

ABSTRACT

Adaptive immune resistance (AIR) is a protective process used by cancer to escape elimination by CD8+ T cells. Inhibition of immune checkpoints PD-1 and CTLA-4 specifically target Interferon-gamma (IFNγ)-driven AIR. AIR begins at the plasma membrane where tumor cell-intrinsic cytokine signaling is initiated. Thus, plasma membrane remodeling by endomembrane trafficking could regulate AIR. Herein we report that the trafficking protein ADP-Ribosylation Factor 6 (ARF6) is critical for IFNγ-driven AIR. ARF6 prevents transport of the receptor to the lysosome, augmenting IFNγR expression, tumor intrinsic IFNγ signaling and downstream expression of immunosuppressive genes. In murine melanoma, loss of ARF6 causes resistance to immune checkpoint blockade (ICB). Likewise, low expression of ARF6 in patient tumors correlates with inferior outcomes with ICB. Our data provide new mechanistic insights into tumor immune escape, defined by ARF6-dependent AIR, and support that ARF6-dependent endomembrane trafficking of the IFNγ receptor influences outcomes of ICB.

3.
Neuron ; 110(19): 3106-3120.e7, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35961320

ABSTRACT

Breakdown of the blood-central nervous system barrier (BCNSB) is a hallmark of many neuroinflammatory disorders, such as multiple sclerosis (MS). Using a mouse model of MS, experimental autoimmune encephalomyelitis (EAE), we show that endothelial-to-mesenchymal transition (EndoMT) occurs in the CNS before the onset of clinical symptoms and plays a major role in the breakdown of BCNSB function. EndoMT can be induced by an IL-1ß-stimulated signaling pathway in which activation of the small GTPase ADP ribosylation factor 6 (ARF6) leads to crosstalk with the activin receptor-like kinase (ALK)-SMAD1/5 pathway. Inhibiting the activation of ARF6 both prevents and reverses EndoMT, stabilizes BCNSB function, reduces demyelination, and attenuates symptoms even after the establishment of severe EAE, without immunocompromising the host. Pan-inhibition of ALKs also reduces disease severity in the EAE model. Therefore, multiple components of the IL-1ß-ARF6-ALK-SMAD1/5 pathway could be targeted for the treatment of a variety of neuroinflammatory disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Monomeric GTP-Binding Proteins , Multiple Sclerosis , Activin Receptors/metabolism , Animals , Central Nervous System/metabolism , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/metabolism , Neuroinflammatory Diseases , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
4.
Transl Vis Sci Technol ; 9(6): 8, 2020 05.
Article in English | MEDLINE | ID: mdl-32821505

ABSTRACT

Purpose: This study aims to develop an impedance-based drug screening platform that will help identify drugs that can enhance the vascular barrier function by stabilizing vascular endothelial cell junctions. Methods: Changes in permeability of cultured human retinal microvascular endothelial cells (HRMECs) monolayer were monitored in real-time with the xCELLigence RTCA system. Using this platform, we performed a primary screen of 2100 known drugs and confirmed hits using two additional secondary permeability assays: the transwell permeability assay and the XPerT assay. The cellular and molecular mechanisms of action and in vivo therapeutic efficacy were also assessed. Results: Eleven compounds blocked interleukin 1 beta (IL-1ß) induced hyperpermeability in the primary screen. Two of 11 compounds, apigenin and ethaverine hydrochloride, reproducibly blocked multiple cytokines induced hyperpermeability. In addition to HRMEC monolayers, the two compounds stabilized three other types of primary vascular endothelial cell monolayers. Preliminary mechanistic studies suggest that the two compounds stabilize the endothelium by blocking ADP-ribosylation factor 6 (ARF6) activation, which results in enhanced VE-cadherin membrane localization. The two compounds showed in vivo efficacy in an animal model of retinal permeability. Conclusions: We developed an impedance-based cellular phenotypic drug screening platform that can identify drugs that enhance vascular barrier function. We found apigenin and ethaverine hydrochloride stabilize endothelial cell junctions and enhance the vascular barrier by blocking ARF6 activation and increasing VE-cadherin membrane localization. Translational Relevance: The drugs identified from the phenotypic screen would have potential therapeutic efficacy in retinal vascular diseases regardless of the underlying mechanisms that promote vascular leak.


Subject(s)
Capillary Permeability , Endothelial Cells , ADP-Ribosylation Factor 6 , Animals , Apigenin/pharmacology , Blood-Retinal Barrier , Humans , Papaverine/analogs & derivatives
5.
Article in English | MEDLINE | ID: mdl-32393494

ABSTRACT

The rise in multidrug-resistant (MDR) organisms portends a serious global threat to the health care system with nearly untreatable infectious diseases, including pneumonia and its often fatal sequelae, acute respiratory distress syndrome (ARDS) and sepsis. Gram-negative bacteria (GNB), including Acinetobacter baumannii, Pseudomonas aeruginosa, and carbapenemase-producing Klebsiella pneumoniae (CPKP), are among the World Health Organization's and National Institutes of Health's high-priority MDR pathogens for targeted development of new therapies. Here, we show that stabilizing the host's vasculature by genetic deletion or pharmacological inhibition of the small GTPase ADP-ribosylation factor 6 (ARF6) increases survival rates of mice infected with A. baumannii, P. aeruginosa, and CPKP. We show that the pharmacological inhibition of ARF6-GTP phenocopies endothelium-specific Arf6 disruption in enhancing the survival of mice with A. baumannii pneumonia, suggesting that inhibition is on target. Finally, we show that the mechanism of protection elicited by these small-molecule inhibitors acts by the restoration of vascular integrity disrupted by GNB lipopolysaccharide (LPS) activation of the TLR4/MyD88/ARNO/ARF6 pathway. By targeting the host's vasculature with small-molecule inhibitors of ARF6 activation, we circumvent microbial drug resistance and provide a potential alternative/adjunctive treatment for emerging and reemerging pathogens.


Subject(s)
Acinetobacter baumannii , Gram-Negative Bacterial Infections , ADP-Ribosylation Factor 6 , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial/genetics , Gram-Negative Bacteria , Gram-Negative Bacterial Infections/drug therapy , Mice , Microbial Sensitivity Tests , Pseudomonas aeruginosa
6.
J Clin Invest ; 127(12): 4569-4582, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29058688

ABSTRACT

The devastating sequelae of diabetes mellitus include microvascular permeability, which results in retinopathy. Despite clinical and scientific advances, there remains a need for new approaches to treat retinopathy. Here, we have presented a possible treatment strategy, whereby targeting the small GTPase ARF6 alters VEGFR2 trafficking and reverses signs of pathology in 4 animal models that represent features of diabetic retinopathy and in a fifth model of ocular pathological angiogenesis. Specifically, we determined that the same signaling pathway utilizes distinct GEFs to sequentially activate ARF6, and these GEFs exert distinct but complementary effects on VEGFR2 trafficking and signal transduction. ARF6 activation was independently regulated by 2 different ARF GEFs - ARNO and GEP100. Interaction between VEGFR2 and ARNO activated ARF6 and stimulated VEGFR2 internalization, whereas a VEGFR2 interaction with GEP100 activated ARF6 to promote VEGFR2 recycling via coreceptor binding. Intervening in either pathway inhibited VEGFR2 signal output. Finally, using a combination of in vitro, cellular, genetic, and pharmacologic techniques, we demonstrated that ARF6 is pivotal in VEGFR2 trafficking and that targeting ARF6-mediated VEGFR2 trafficking has potential as a therapeutic approach for retinal vascular diseases such as diabetic retinopathy.


Subject(s)
ADP-Ribosylation Factors/metabolism , Diabetic Retinopathy/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Cell Line , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Protein Transport , Vascular Endothelial Growth Factor Receptor-2/genetics
7.
Cancer Cell ; 29(6): 889-904, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27265506

ABSTRACT

Activating mutations in Gαq proteins, which form the α subunit of certain heterotrimeric G proteins, drive uveal melanoma oncogenesis by triggering multiple downstream signaling pathways, including PLC/PKC, Rho/Rac, and YAP. Here we show that the small GTPase ARF6 acts as a proximal node of oncogenic Gαq signaling to induce all of these downstream pathways as well as ß-catenin signaling. ARF6 activates these diverse pathways through a common mechanism: the trafficking of GNAQ and ß-catenin from the plasma membrane to cytoplasmic vesicles and the nucleus, respectively. Blocking ARF6 with a small-molecule inhibitor reduces uveal melanoma cell proliferation and tumorigenesis in a mouse model, confirming the functional relevance of this pathway and suggesting a therapeutic strategy for Gα-mediated diseases.


Subject(s)
ADP-Ribosylation Factors/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Melanoma/drug therapy , Small Molecule Libraries/administration & dosage , Uveal Neoplasms/drug therapy , beta Catenin/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/antagonists & inhibitors , ADP-Ribosylation Factors/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cytoplasm/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Mice , Neoplasm Transplantation , Protein Transport/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism
8.
Tetrahedron ; 69(27-28): 5744-5750, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23807819

ABSTRACT

Cationic Rh(II) complexes are able to catalyze the regioselective hydroamination of propargyl ureas in a 6-endo fashion. This transformation permits access to interesting substitution patterns of dihydropyrimidines which have found use as nucleotide exchange factor inhibitors.

9.
Sci Signal ; 6(265): ra14, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23462101

ABSTRACT

ß-Catenin has a dual function in cells: fortifying cadherin-based adhesion at the plasma membrane and activating transcription in the nucleus. We found that in melanoma cells, WNT5A stimulated the disruption of N-cadherin and ß-catenin complexes by activating the guanosine triphosphatase adenosine diphosphate ribosylation factor 6 (ARF6). Binding of WNT5A to the Frizzled 4-LRP6 (low-density lipoprotein receptor-related protein 6) receptor complex activated ARF6, which liberated ß-catenin from N-cadherin, thus increasing the pool of free ß-catenin, enhancing ß-catenin-mediated transcription, and stimulating invasion. In contrast to WNT5A, the guidance cue SLIT2 and its receptor ROBO1 inhibited ARF6 activation and, accordingly, stabilized the interaction of N-cadherin with ß-catenin and reduced transcription and invasion. Thus, ARF6 integrated competing signals in melanoma cells, thereby enabling plasticity in the response to external cues. Moreover, small-molecule inhibition of ARF6 stabilized adherens junctions, blocked ß-catenin signaling and invasiveness of melanoma cells in culture, and reduced spontaneous pulmonary metastasis in mice, suggesting that targeting ARF6 may provide a means of inhibiting WNT/ß-catenin signaling in cancer.


Subject(s)
ADP-Ribosylation Factors/physiology , Melanoma/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins/physiology , Transcriptional Activation/physiology , Wnt Proteins/physiology , beta Catenin/physiology , ADP-Ribosylation Factor 6 , Gene Silencing , Humans , Signal Transduction , Wnt-5a Protein , beta Catenin/metabolism
10.
Proc Natl Acad Sci U S A ; 110(13): 5181-6, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479632

ABSTRACT

Stargardt type 3 (STGD3) disease is a juvenile macular dystrophy caused by mutations in the ELOVL4 (Elongation of very long chain fatty acids 4) gene. Its protein product, ELOVL4, is an elongase required for the biosynthesis of very long-chain polyunsaturated fatty acids (VLC-PUFAs). It is unclear whether photoreceptor degeneration in STGD3 is caused by loss of VLC-PUFAs or by mutated ELOVL4 protein trafficking/aggregation. We therefore generated conditional knockout (cKO) mice with Elovl4 ablated in rods or cones and compared their phenotypes to transgenic (TG) animals that express the human STGD3-causing ELOVL4(STGD3) allele. Gas chromatography-mass spectrometry was used to assess C30-C34 VLC-PUFA and N-retinylidene-N-retinylethanolamine content; electroretinography was used to measure phototransduction and outer retinal function; electron microscopy was used for retinal ultrastructure; and the optomotor tracking response was used to test scotopic and photopic visual performance. Elovl4 transcription and biosynthesis of C30-C34 VLC-PUFAs in rod cKO and TG retinas were reduced up to 98%, whereas the content of docosahexaenoic acid was diminished in TG, but not rod cKO, retinas. Despite the near-total loss of the retinal VLC-PUFA content, rod and cone cKO animals exhibited no electrophysiological or behavioral deficits, whereas the typical rod-cone dystrophic pattern was observed in TG animals. Our data suggest that photoreceptor-specific VLC-PUFA depletion is not sufficient to induce the STGD3 phenotype, because depletion alone had little effect on photoreceptor survival, phototransduction, synaptic transmission, and visual behavior.


Subject(s)
Eye Proteins/metabolism , Fatty Acids, Unsaturated/metabolism , Macular Degeneration/congenital , Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Synaptic Transmission , Vision, Ocular , Animals , Cell Survival/genetics , Disease Models, Animal , Eye Proteins/genetics , Fatty Acids, Unsaturated/genetics , Humans , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Photoreceptor Cells, Vertebrate/pathology , Retina/pathology
11.
Nature ; 492(7428): 252-5, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23143332

ABSTRACT

The innate immune response is essential for combating infectious disease. Macrophages and other cells respond to infection by releasing cytokines, such as interleukin-1ß (IL-1ß), which in turn activate a well-described, myeloid-differentiation factor 88 (MYD88)-mediated, nuclear factor-κB (NF-κB)-dependent transcriptional pathway that results in inflammatory-cell activation and recruitment. Endothelial cells, which usually serve as a barrier to the movement of inflammatory cells out of the blood and into tissue, are also critical mediators of the inflammatory response. Paradoxically, the cytokines vital to a successful immune defence also have disruptive effects on endothelial cell-cell interactions and can trigger degradation of barrier function and dissociation of tissue architecture. The mechanism of this barrier dissolution and its relationship to the canonical NF-κB pathway remain poorly defined. Here we show that the direct, immediate and disruptive effects of IL-1ß on endothelial stability in a human in vitro cell model are NF-κB independent and are instead the result of signalling through the small GTPase ADP-ribosylation factor 6 (ARF6) and its activator ARF nucleotide binding site opener (ARNO; also known as CYTH2). Moreover, we show that ARNO binds directly to the adaptor protein MYD88, and thus propose MYD88-ARNO-ARF6 as a proximal IL-1ß signalling pathway distinct from that mediated by NF-κB. Finally, we show that SecinH3, an inhibitor of ARF guanine nucleotide-exchange factors such as ARNO, enhances vascular stability and significantly improves outcomes in animal models of inflammatory arthritis and acute inflammation.


Subject(s)
ADP-Ribosylation Factors/metabolism , GTPase-Activating Proteins/metabolism , Myeloid Differentiation Factor 88/metabolism , Receptors, Interleukin/metabolism , ADP-Ribosylation Factor 6 , Adjuvants, Immunologic/pharmacology , Animals , Arthritis/pathology , Cadherins/metabolism , Capillary Permeability/drug effects , Cell Line , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Humans , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Purines/pharmacology , Signal Transduction , Thiophenes/pharmacology
12.
J Biol Chem ; 287(14): 11469-80, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22199362

ABSTRACT

Very long chain polyunsaturated fatty acid (VLC-PUFA)-containing glycerophospholipids are highly enriched in the retina; however, details regarding the specific synthesis and function of these highly unusual retinal glycerophospholipids are lacking. Elongation of very long chain fatty acids-4 (ELOVL4) has been identified as a fatty acid elongase protein involved in the synthesis of VLC-PUFAs. Mutations in ELOVL4 have also been implicated in an autosomal dominant form of Stargardt disease (STGD3), a type of juvenile macular degeneration. We have generated photoreceptor-specific conditional knock-out mice and used high performance liquid chromatography-mass spectrometry (HPLC-MS) to examine and analyze the fatty acid composition of retinal membrane glycerophosphatidylcholine and glycerophosphatidylethanolamine species. We also used immunofluorescent staining and histology coupled with electrophysiological data to assess retinal morphology and visual response. The conditional knock-out mice showed a significant decrease in retinal glycerophospholipids containing VLC-PUFAs, specifically contained in the sn-1 position of glycerophosphatidylcholine, implicating the role of Elovl4 in their synthesis. Conditional knock-out mice were also found to have abnormal accumulation of lipid droplets and lipofuscin-like granules while demonstrating photoreceptor-specific abnormalities in visual response, indicating the critical role of Elovl4 for proper rod or cone photoreceptor function. Altogether, this study demonstrates the essential role of ELOVL4 in VLC-PUFA synthesis and retinal function.


Subject(s)
Eye Proteins/metabolism , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids, Unsaturated/chemistry , Membrane Proteins/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Animals , Cell Membrane/metabolism , Electrophysiological Phenomena , Eye Proteins/genetics , Gene Knockout Techniques , Glycerophospholipids/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Phosphorylcholine/metabolism
13.
Proc Natl Acad Sci U S A ; 108(35): 14578-83, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21844367

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in the elderly. Wet AMD includes typical choroidal neovascularization (CNV) and polypoidal choroidal vasculopathy (PCV). The etiology and pathogenesis of CNV and PCV are not well understood. Genome-wide association studies have linked a multifunctional serine protease, HTRA1, to AMD. However, the precise role of HTRA1 in AMD remains elusive. By transgenically expressing human HTRA1 in mouse retinal pigment epithelium, we showed that increased HTRA1 induced cardinal features of PCV, including branching networks of choroidal vessels, polypoidal lesions, severe degeneration of the elastic laminae, and tunica media of choroidal vessels. In addition, HTRA1 mice displayed retinal pigment epithelium atrophy and photoreceptor degeneration. Senescent HTRA1 mice developed occult CNV, which likely resulted from the degradation of the elastic lamina of Bruch's membrane and up-regulation of VEGF. Our results indicate that increased HTRA1 is sufficient to cause PCV and is a significant risk factor for CNV.


Subject(s)
Macular Degeneration/etiology , Neovascularization, Pathologic , Retinal Pigment Epithelium/enzymology , Serine Endopeptidases/physiology , Animals , Choroidal Neovascularization/etiology , High-Temperature Requirement A Serine Peptidase 1 , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pancreatic Elastase/metabolism , Proteins/physiology , Vascular Endothelial Growth Factor A/physiology
14.
Mol Vis ; 16: 2273-8, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21139680

ABSTRACT

PURPOSE: Age-related macular degeneration (AMD) is the most common cause of irreversible central vision loss worldwide. Research has linked AMD susceptibility with dysregulation of the complement cascade. Typically, complement factor H (CFH), complement factor B (CFB), complement component 2 (C2), and complement component 3 (C3) are associated with AMD. In this paper, we investigated the association between complement factor D (CFD), another factor of the complement system, and advanced AMD in a Caucasian population. METHODS: Six single nucleotide polymorphisms (SNPs), rs1683564, rs35186399, rs1683563, rs3826945, rs34337649, and rs1651896, across the region covering CFD, were chosen for this study. One hundred and seventy-eight patients with advanced AMD and 161 age-matched normal controls were genotyped. Potential positive signals were further tested in another independent 445 advanced AMD patients and 190 controls. χ2 tests were performed to compare the allele frequencies between case and control groups. RESULTS: None of the six SNPs of CFD was found to be significantly associated with advanced AMD in our study. CONCLUSIONS: Our findings suggest that CFD may not play a major role in the genetic susceptibility to AMD because no association was found between the six SNPs analyzed in the CFD region and advanced AMD.


Subject(s)
Complement Factor D/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Macular Degeneration/genetics , Macular Degeneration/pathology , Polymorphism, Single Nucleotide/genetics , Base Sequence , Complement Pathway, Alternative/genetics , Humans , Linkage Disequilibrium/genetics , Middle Aged , Molecular Sequence Data
15.
Mol Vis ; 16: 2062-70, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-21042561

ABSTRACT

PURPOSE: Two Chinese families (XT and YT) with congenital fibrosis of the extraocular muscles (CFEOM) were identified. The purpose of this study was to determine if previously described Homo sapiens kinesin family member 21A (KIF21A) mutations were responsible for CFEOM in these two Chinese pedigrees. METHODS: Clinical characterization and genetic studies were performed. Microsatellite genotyping for linkage to the CFEOM1 and CFEOM3 loci was performed. The probands were screened for KIF21A mutations by bidirectional direct sequencing. Once a mutation was detected in the proband, all other participating family members and 100 unrelated control normal individuals were screened for the mutation. RESULTS: All affected individuals in family XT shared the common manifestations of CFEOM1. Family YT had two affected individuals, a mother and a daughter. The daughter had CFEOM1, while her mother never had congential ptosis but did have limited extraocular movements status post strabismus surgery. Haplotype analysis revealed that pedigree XT was linked to the 12q CFEOM1 locus and the affected memberes harbored the second most common missense mutation in KIF21A (2,861G>A, R954Q). Family YT harbored the most common missense de novo mutation in KIF21A (2,860C>T, R954W). Both of these mutations have been previously described. CONCLUSIONS: The observation of these two KIF21A mutations in a Chinese pedigree underscores the homogeneity of these mutations as a cause of CFEOM1 and CFEOM3 across ethnic divisions.


Subject(s)
Asian People/genetics , Kinesins/genetics , Mutation/genetics , Ocular Motility Disorders/congenital , Ocular Motility Disorders/genetics , Oculomotor Muscles/pathology , Adolescent , Base Sequence , China , DNA Mutational Analysis , Eye Movements/genetics , Family , Female , Fibrosis , Haplotypes/genetics , Humans , Lod Score , Male , Microsatellite Repeats/genetics , Molecular Sequence Data , Ocular Motility Disorders/physiopathology , Oculomotor Muscles/physiopathology , Pedigree
16.
PLoS Genet ; 6(2): e1000836, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20140183

ABSTRACT

A common haplotype on 10q26 influences the risk of age-related macular degeneration (AMD) and encompasses two genes, LOC387715 and HTRA1. Recent data have suggested that loss of LOC387715, mediated by an insertion/deletion (in/del) that destabilizes its message, is causally related with the disorder. Here we show that loss of LOC387715 is insufficient to explain AMD susceptibility, since a nonsense mutation (R38X) in this gene that leads to loss of its message resides in a protective haplotype. At the same time, the common disease haplotype tagged by the in/del and rs11200638 has an effect on the transcriptional upregulation of the adjacent gene, HTRA1. These data implicate increased HTRA1 expression in the pathogenesis of AMD and highlight the importance of exploring multiple functional consequences of alleles in haplotypes that confer susceptibility to complex traits.


Subject(s)
Macular Degeneration/genetics , Proteins/genetics , Serine Endopeptidases/genetics , Aged , Case-Control Studies , Chromosomes, Human, Pair 10/genetics , Cohort Studies , Enzyme Assays , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Haplotypes/genetics , High-Temperature Requirement A Serine Peptidase 1 , Humans , Luciferases/metabolism , Male , Polymorphism, Single Nucleotide/genetics , Proteins/metabolism , Serine Endopeptidases/metabolism , Utah
17.
Proc Natl Acad Sci U S A ; 106(40): 17105-10, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19805132

ABSTRACT

Primary open-angle glaucoma (POAG) is the second leading cause of blindness worldwide. Although a number of genetic loci have shown association or genetic linkage to monogenic forms of POAG, the identified genes and loci do not appear to have a major role in the common POAG phenotype. We seek to identify genetic loci that appear to be major risk factors for POAG in the Afro-Caribbean population of Barbados, West Indies. We performed linkage analyses in 146 multiplex families ascertained through the Barbados Family Study of Glaucoma (BFSG) and identified a strong linkage signal on chromosome 2p (logarithm of odds score = 6.64 at = 0 with marker D2S2156). We subsequently performed case-control analyses using unrelated affected individuals and unaffected controls. A set of SNPs on chromosome 2p was evaluated in two independent groups of BFSG participants, a discovery group (130 POAG cases, 65 controls) and a replication group (122 POAG cases, 65 controls), and a strong association was identified with POAG and rs12994401 in both groups (P < 3.34 E-09 and P < 1.21E-12, respectively). The associated SNPs form a common disease haplotype. In summary, we have identified a locus with a major impact on susceptibility to the common POAG phenotype in an Afro-Caribbean population in Barbados. Our approach illustrates the merit of using an isolated population enriched with common disease variants as an efficient method to identify genetic underpinning of POAG.


Subject(s)
Chromosomes, Human, Pair 2/genetics , Glaucoma, Open-Angle/genetics , Polymorphism, Single Nucleotide , Barbados , Black People/ethnology , Black People/genetics , Case-Control Studies , Family Health , Female , Gene Frequency , Genetic Variation , Genotype , Glaucoma, Open-Angle/ethnology , Haplotypes , Humans , Male , Risk Factors
18.
N Engl J Med ; 359(14): 1456-63, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18753640

ABSTRACT

BACKGROUND: Age-related macular degeneration is the most common cause of irreversible visual impairment in the developed world. Advanced age-related macular degeneration consists of geographic atrophy and choroidal neovascularization. The specific genetic variants that predispose patients to geographic atrophy are largely unknown. METHODS: We tested for an association between the functional toll-like receptor 3 gene (TLR3) variant rs3775291 (involving the substitution of phenylalanine for leucine at amino acid 412) and age-related macular degeneration in Americans of European descent. We also tested for the effect of TLR3 Leu and Phe variants on the viability of human retinal pigment epithelial cells in vitro and on apoptosis of retinal pigment epithelial cells from wild-type mice and Tlr3-knockout (Tlr3(-/-)) mice. RESULTS: The Phe variant (encoded by the T allele at rs3775291) was associated with protection against geographic atrophy (P=0.005). This association was replicated in two independent case-control series of geographic atrophy (P=5.43x10(-4) and P=0.002). No association was found between TLR3 variants and choroidal neovascularization. A prototypic TLR3 ligand induced apoptosis in a greater fraction of human retinal pigment epithelial cells with the Leu-Leu genotype than those with the Leu-Phe genotype and in a greater fraction of wild-type mice than Tlr3(-/-) mice. CONCLUSIONS: The TLR3 412Phe variant confers protection against geographic atrophy, probably by suppressing the death of retinal pigment epithelial cells. Since double-stranded RNA (dsRNA) can activate TLR3-mediated apoptosis, our results suggest a role of viral dsRNA in the development of geographic atrophy and point to the potential toxic effects of short-interfering-RNA therapies in the eye.


Subject(s)
Macula Lutea/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Toll-Like Receptor 3/genetics , Animals , Apoptosis , Case-Control Studies , Choroidal Neovascularization/genetics , Genotype , Humans , In Vitro Techniques , Interferon Inducers/pharmacology , Mice , Mice, Knockout , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/drug effects , Pigment Epithelium of Eye/pathology , Poly I-C/pharmacology , Polymorphism, Single Nucleotide , RNA, Double-Stranded/adverse effects , RNA, Small Interfering/adverse effects , RNA, Viral/adverse effects
19.
Proc Natl Acad Sci U S A ; 105(19): 6998-7003, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18458324

ABSTRACT

Significant morbidity and mortality among patients with diabetes mellitus result largely from a greatly increased incidence of microvascular complications. Proliferative diabetic retinopathy (PDR) and end stage renal disease (ESRD) are two of the most common and severe microvascular complications of diabetes. A high concordance exists in the development of PDR and ESRD in diabetic patients, as well as strong familial aggregation of these complications, suggesting a common underlying genetic mechanism. However, the precise gene(s) and genetic variant(s) involved remain largely unknown. Erythropoietin (EPO) is a potent angiogenic factor observed in the diabetic human and mouse eye. By a combination of case-control association and functional studies, we demonstrate that the T allele of SNP rs1617640 in the promoter of the EPO gene is significantly associated with PDR and ESRD in three European-American cohorts [Utah: P = 1.91 x 10(-3); Genetics of Kidneys in Diabetes (GoKinD) Study: P = 2.66 x 10(-8); and Boston: P = 2.1 x 10(-2)]. The EPO concentration in human vitreous body was 7.5-fold higher in normal subjects with the TT risk genotype than in those with the GG genotype. Computational analysis suggests that the risk allele (T) of rs1617640 creates a matrix match with the EVI1/MEL1 or AP1 binding site, accounting for an observed 25-fold enhancement of luciferase reporter expression as compared with the G allele. These results suggest that rs1617640 in the EPO promoter is significantly associated with PDR and ESRD. This study identifies a disease risk-associated gene and potential pathway mediating severe diabetic microvascular complications.


Subject(s)
Diabetic Nephropathies/complications , Diabetic Nephropathies/genetics , Diabetic Retinopathy/complications , Diabetic Retinopathy/genetics , Erythropoietin/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Alleles , Animals , Cell Line , Cohort Studies , Erythropoietin/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Predisposition to Disease , Haplotypes , Humans , Kidney/metabolism , Kidney/pathology , Luciferases/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology
20.
Cell Cycle ; 7(4): 521-4, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18287813

ABSTRACT

Exfoliation glaucoma (XFG) is the commonest identifiable cause of secondary open-angle glaucoma worldwide, characterized by the deposition of fibrillar proteins in the anterior segment of the eye. We investigated LOXL1 gene variants previously identified to confer susceptibility to XFG in a Utah Caucasian cohort. After a standard eye examination protocol we genotyped SNPs rs2165241and rs3825942 in 62 XFG or exfoliation syndrome (XFS) patients and 170 normal controls. Genotype frequency distribution, odds ratios (ORs) and population attributable risks were calculated for the risk alleles. The SNP rs2165241 was significantly associated with XFG and XFS (p = 4.13 x 10(-9)) for an additive model, OR(het) = 4.42 (2.30-8.50), OR(hom) = 34.19 (4.48-261.00); T allele: 83.1% in cases versus 52.4% in controls). Significant association was also found for rs3825942: (p = 1.89 x 10(-6)). Our findings confirm genetic association of LOXL1 with XFG and XFS and implicate a potential role of cross linking of elastin in the pathogenesis of XFG. This information will potentially guide glaucoma monitoring efforts by targeting individuals whose genetic profiles put them at higher risk for XFG.


Subject(s)
Amino Acid Oxidoreductases/genetics , Elastin/metabolism , Exfoliation Syndrome/complications , Exfoliation Syndrome/genetics , Genetic Predisposition to Disease , Glaucoma, Open-Angle/etiology , Cohort Studies , DNA Primers/genetics , Female , Genotype , Glaucoma, Open-Angle/genetics , Humans , Male , Odds Ratio , Polymorphism, Single Nucleotide , Utah , White People
SELECTION OF CITATIONS
SEARCH DETAIL