Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Crit Rev Biochem Mol Biol ; 55(4): 372-385, 2020 08.
Article in English | MEDLINE | ID: mdl-32727223

ABSTRACT

The Eyes Absent (EYA) proteins are the only known instance of a single polypeptide housing the following three separable biochemical activities: tyrosine phosphatase, threonine phosphatase, and transactivation. This uniquely positions the EYAs to participate in both transcriptional regulation and signal transduction pathways. But it also complicates the assignment of biological roles to individual biochemical activities through standard loss-of-function experiments. Nevertheless, there is an emerging literature linking developmental and pathological functions with the various EYA activities, and a growing list of disease states that might benefit from EYA-targeted therapeutics. There also remain multiple unresolved issues with significant implications for our understanding of how the EYAs might impact such ubiquitous signaling cascades as the MYC and Notch pathways. This review will describe the unique juxtaposition of biochemical activities in the EYAs, their interaction with signaling pathways and cellular processes, emerging evidence of roles in disease states, and the feasibility of therapeutic targeting of individual EYA activities. We will focus on the phosphatase activities of the vertebrate EYA proteins and will examine the current state of knowledge regarding: • substrates and signaling pathways affected by the EYA tyrosine phosphatase activity; • modes of regulation of the EYA tyrosine phosphatase activity; • signaling pathways that implicate the threonine phosphatase activity of the EYAs including a potential interaction with PP2A-B55α; • the interplay between the two phosphatase activities and the transactivation function of the EYAs; • disease states associated with the EYAs and the current state of development of EYA-targeted therapeutics.


Subject(s)
Phosphoprotein Phosphatases/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Humans , Phosphoprotein Phosphatases/genetics , Protein Tyrosine Phosphatases/genetics , Trans-Activators/genetics
2.
Circulation ; 144(7): 539-555, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34111939

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a common complication in patients with alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a severe congenital disorder associated with mutations in the FOXF1 gene. Although the loss of alveolar microvasculature causes PH in patients with ACDMPV, it is unknown whether increasing neonatal lung angiogenesis could prevent PH and right ventricular (RV) hypertrophy. METHODS: We used echocardiography, RV catheterization, immunostaining, and biochemical methods to examine lung and heart remodeling and RV output in Foxf1WT/S52F mice carrying the S52F Foxf1 mutation (identified in patients with ACDMPV). The ability of Foxf1WT/S52F mutant embryonic stem cells to differentiate into respiratory cell lineages in vivo was examined using blastocyst complementation. Intravascular delivery of nanoparticles with a nonintegrating Stat3 expression vector was used to improve neonatal pulmonary angiogenesis in Foxf1WT/S52F mice and determine its effects on PH and RV hypertrophy. RESULTS: Foxf1WT/S52F mice developed PH and RV hypertrophy after birth. The severity of PH in Foxf1WT/S52F mice directly correlated with mortality, low body weight, pulmonary artery muscularization, and increased collagen deposition in the lung tissue. Increased fibrotic remodeling was found in human ACDMPV lungs. Mouse embryonic stem cells carrying the S52F Foxf1 mutation were used to produce chimeras through blastocyst complementation and to demonstrate that Foxf1WT/S52F embryonic stem cells have a propensity to differentiate into pulmonary myofibroblasts. Intravascular delivery of nanoparticles carrying Stat3 cDNA protected Foxf1WT/S52F mice from RV hypertrophy and PH, improved survival, and decreased fibrotic lung remodeling. CONCLUSIONS: Nanoparticle therapies increasing neonatal pulmonary angiogenesis may be considered to prevent PH in ACDMPV.


Subject(s)
Gene Transfer Techniques , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/therapy , Nanoparticles , Persistent Fetal Circulation Syndrome/complications , Pulmonary Alveoli/abnormalities , STAT3 Transcription Factor/genetics , Airway Remodeling/genetics , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Drug Carriers , Drug Delivery Systems , Echocardiography , Fibrosis , Forkhead Transcription Factors/deficiency , Genetic Therapy , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/diagnosis , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Mice , Mice, Transgenic , Microvascular Density/genetics , Myofibroblasts/metabolism , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , STAT3 Transcription Factor/administration & dosage , Theranostic Nanomedicine/methods , Treatment Outcome , Vascular Remodeling/genetics
3.
Biochem Soc Trans ; 49(3): 1397-1408, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34196366

ABSTRACT

The Eyes Absent (EYA) transactivator-phosphatase proteins are important contributors to cell-fate determination processes and to the development of multiple organs. The transcriptional regulatory activity as well as the protein tyrosine phosphatase activities of the EYA proteins can independently contribute to proliferation, differentiation, morphogenesis and tissue homeostasis in different contexts. Aberrant EYA levels or activity are associated with numerous syndromic and non-syndromic developmental disorders, as well as cancers. Commensurate with the multiplicity of biochemical activities carried out by the EYA proteins, they impact upon a range of cellular signaling pathways. Here, we provide a broad overview of the roles played by EYA proteins in development, and highlight the molecular signaling pathways known to be linked with EYA-associated organ development and developmental disorders.


Subject(s)
Congenital Abnormalities/genetics , Eye/metabolism , Gene Expression Regulation, Developmental , Kidney/metabolism , Protein Tyrosine Phosphatases/genetics , Trans-Activators/genetics , Animals , Congenital Abnormalities/embryology , Congenital Abnormalities/metabolism , Eye/embryology , Eye/growth & development , Genetic Predisposition to Disease/genetics , Humans , Kidney/embryology , Kidney/growth & development , Mutation , Protein Tyrosine Phosphatases/metabolism , Trans-Activators/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920226

ABSTRACT

Here, we review the haloacid dehalogenase (HAD) class of protein phosphatases, with a particular emphasis on an unusual group of enzymes, the eyes absent (EYA) family. EYA proteins have the unique distinction of being structurally and mechanistically classified as HAD enzymes, yet, unlike other HAD phosphatases, they are protein tyrosine phosphatases (PTPs). Further, the EYA proteins are unique among the 107 classical PTPs in the human genome because they do not use a Cysteine residue as a nucleophile in the dephosphorylation reaction. We will provide an overview of HAD phosphatase structure-function, describe unique features of the EYA family and their tyrosine phosphatase activity, provide a brief summary of the known substrates and cellular functions of the EYA proteins, and speculate about the evolutionary origins of the EYA family of proteins.


Subject(s)
Evolution, Molecular , Genome, Human/genetics , Hydrolases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Cysteine/metabolism , Humans , Hydrolases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Structure-Activity Relationship
5.
Nature ; 494(7436): 243-6, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23334418

ABSTRACT

Vascular patterning is critical for organ function. In the eye, there is simultaneous regression of embryonic hyaloid vasculature (important to clear the optical path) and formation of the retinal vasculature (important for the high metabolic demands of retinal neurons). These events occur postnatally in the mouse. Here we have identified a light-response pathway that regulates both processes. We show that when mice are mutated in the gene (Opn4) for the atypical opsin melanopsin, or are dark-reared from late gestation, the hyaloid vessels are persistent at 8 days post-partum and the retinal vasculature overgrows. We provide evidence that these vascular anomalies are explained by a light-response pathway that suppresses retinal neuron number, limits hypoxia and, as a consequence, holds local expression of vascular endothelial growth factor (VEGFA) in check. We also show that the light response for this pathway occurs in late gestation at about embryonic day 16 and requires the photopigment in the fetus and not the mother. Measurements show that visceral cavity photon flux is probably sufficient to activate melanopsin-expressing retinal ganglion cells in the mouse fetus. These data thus show that light--the stimulus for function of the mature eye--is also critical in preparing the eye for vision by regulating retinal neuron number and initiating a series of events that ultimately pattern the ocular blood vessels.


Subject(s)
Eye/blood supply , Eye/growth & development , Fetus/radiation effects , Light Signal Transduction/radiation effects , Light , Retinal Neurons/radiation effects , Rod Opsins/metabolism , Animals , Cell Count , Cell Hypoxia/radiation effects , Eye/metabolism , Eye/radiation effects , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Neovascularization, Physiologic/radiation effects , Photons , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Retinal Neurons/cytology , Retinal Neurons/metabolism , Rod Opsins/deficiency , Rod Opsins/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Angiogenesis ; 20(3): 307-323, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28108843

ABSTRACT

ETS transcription factor ETV2/Etsrp functions as a key regulator of embryonic vascular development in multiple vertebrates. However, its role in pathological vascular development has not been previously investigated. To analyze its role in tumor angiogenesis, we utilized a zebrafish xenotransplantation model. Using a photoconvertible kdrl:NLS-KikGR line, we demonstrated that all tumor vessels originate from the existing embryonic vasculature by the mechanism of angiogenesis. Xenotransplantation of mouse B16 melanoma cells resulted in a significant increase in expression of the ETS transcription factors etv2 and fli1b expression throughout the embryonic vasculature. etv2 null mutants which undergo significant recovery of embryonic angiogenesis during later developmental stages displayed a strong inhibition of tumor angiogenesis. We utilized highly specific and fully validated photoactivatable morpholinos to inhibit Etv2 function after embryonic vasculogenesis has completed. Inducible inhibition of Etv2 function resulted in a significant reduction of tumor angiogenesis and inhibition of tumor growth. Furthermore, inducible inhibition of Etv2 function in fli1b mutant embryos resulted in even stronger reduction in tumor angiogenesis and growth, demonstrating that Etv2 and Fli1b have a partially redundant requirement during tumor angiogenesis. These results demonstrate the requirement for Etv2 and Fli1b in tumor angiogenesis and suggest that inhibition of these ETS factors may present a novel strategy to inhibit tumor angiogenesis and reduce tumor growth.


Subject(s)
Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Melanoma, Experimental/pathology , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Transcription Factors/genetics , Up-Regulation/genetics , Xenograft Model Antitumor Assays , Zebrafish/embryology , Zebrafish Proteins/genetics
7.
Am J Pathol ; 186(3): 568-78, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26765957

ABSTRACT

Management of neoangiogenesis remains a high-value therapeutic goal. A recently uncovered association between the DNA damage repair pathway and pathological angiogenesis could open previously unexplored possibilities for intervention. An attractive and novel target is the Eyes absent (EYA) tyrosine phosphatase, which plays a critical role in the repair versus apoptosis decision after DNA damage. This study examines the role of EYA in the postnatal development of the retinal vasculature and under conditions of ischemia-reperfusion encountered in proliferative retinopathies. We find that the ability of the EYA proteins to promote endothelial cell (EC) migration contributes to a delay in postnatal development of the retinal vasculature when Eya3 is deleted specifically in ECs. By using genetic and chemical biology tools, we show that EYA contributes to pathological angiogenesis in a model of oxygen-induced retinopathy. Both in vivo and in vitro, loss of EYA tyrosine phosphatase activity leads to defective assembly of γ-H2AX foci and thus to DNA damage repair in ECs under oxidative stress. These data reveal the potential utility of EYA tyrosine phosphatase inhibitors as therapeutic agents in inhibiting pathological neovascularization with a range of clinical applications.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Neovascularization, Pathologic/pathology , Protein Tyrosine Phosphatases/metabolism , Animals , Animals, Newborn , Cell Movement , DNA Damage/physiology , DNA-Binding Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Eye/metabolism , Eye/pathology , Female , Gene Deletion , Humans , Male , Mice, Inbred C57BL , Neovascularization, Pathologic/prevention & control , Oxygen/adverse effects , Protein Tyrosine Phosphatases/genetics , Reperfusion Injury/pathology , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/pathology
8.
BMC Cancer ; 17(1): 338, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28521819

ABSTRACT

BACKGROUND: Multicellular Tumor Spheroids are frequently used to mimic the regionalization of proliferation and the hypoxic environment within avascular tumors. Here we exploit these features to study the activation of DNA damage repair pathways and their correlation to developing hypoxia. METHODS: Activation of DNA damage repair markers, proliferation, cell death, glycogen accumulation and developing hypoxia were investigated using immunofluorescence, immuno-histochemistry, EdU incorporation, Western blots, COMET assays, and pharmacological agents in A673 Ewing sarcoma spheroids and monolayer cultures. RESULTS: DNA damage marker γ-H2AX is observed in the hypoxic, peri-necrotic region of growing spheroids. While most proliferating cells are seen on the spheroid surface, there are also a few Ki-67 positive cells in the hypoxic zone. The hypoxia-induced phosphorylation of H2AX to form γ-H2AX in spheroids is attenuated by the ATM inhibitor KU55933, but not the ATR inhibitor VE-821. CONCLUSION: Tumor spheroids mimic tumor microenvironments such as the anoxic, hypoxic and oxic niches within solid tumors, as well as populations of cells that are viable, proliferating, and undergoing DNA damage repair processes under these different micro-environmental conditions. ATM, but not ATR, is the primary kinase responsible for γ-H2AX formation in the hypoxic core of A673 spheroids. Spheroids could offer unique advantages in testing therapeutics designed to target malignant cells that evade conventional treatment strategies by adapting to the hypoxic tumor microenvironment.


Subject(s)
Cell Proliferation , DNA Damage , Spheroids, Cellular/physiology , Animals , Apoptosis , Carcinoma, Lewis Lung/pathology , Cell Hypoxia , Cell Line, Tumor , Histones/metabolism , Humans , Mice
9.
J Med Genet ; 52(2): 85-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25480986

ABSTRACT

BACKGROUND: Oliver-McFarlane syndrome is characterised by trichomegaly, congenital hypopituitarism and retinal degeneration with choroidal atrophy. Laurence-Moon syndrome presents similarly, though with progressive spinocerebellar ataxia and spastic paraplegia and without trichomegaly. Both recessively inherited disorders have no known genetic cause. METHODS: Whole-exome sequencing was performed to identify the genetic causes of these disorders. Mutations were functionally validated in zebrafish pnpla6 morphants. Embryonic expression was evaluated via in situ hybridisation in human embryonic sections. Human neurohistopathology was performed to characterise cerebellar degeneration. Enzymatic activities were measured in patient-derived fibroblast cell lines. RESULTS: Eight mutations in six families with Oliver-McFarlane or Laurence-Moon syndrome were identified in the PNPLA6 gene, which encodes neuropathy target esterase (NTE). PNPLA6 expression was found in the developing human eye, pituitary and brain. In zebrafish, the pnpla6 curly-tailed morphant phenotype was fully rescued by wild-type human PNPLA6 mRNA and not by mutation-harbouring mRNAs. NTE enzymatic activity was significantly reduced in fibroblast cells derived from individuals with Oliver-McFarlane syndrome. Intriguingly, adult brain histology from a patient with highly overlapping features of Oliver-McFarlane and Laurence-Moon syndromes revealed extensive cerebellar degeneration and atrophy. CONCLUSIONS: Previously, PNPLA6 mutations have been associated with spastic paraplegia type 39, Gordon-Holmes syndrome and Boucher-Neuhäuser syndromes. Discovery of these additional PNPLA6-opathies further elucidates a spectrum of neurodevelopmental and neurodegenerative disorders associated with NTE impairment and suggests a unifying mechanism with diagnostic and prognostic importance.


Subject(s)
Blepharoptosis/enzymology , Blepharoptosis/genetics , Carboxylic Ester Hydrolases/genetics , Dwarfism/enzymology , Dwarfism/genetics , Genetic Predisposition to Disease , Hypertrichosis/enzymology , Hypertrichosis/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Laurence-Moon Syndrome/enzymology , Laurence-Moon Syndrome/genetics , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/genetics , Alleles , Amino Acid Sequence , Animals , Carboxylic Ester Hydrolases/chemistry , Central Nervous System/pathology , Developmental Disabilities/enzymology , Developmental Disabilities/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Humans , Molecular Sequence Data , Mutation/genetics , Phenotype , Phospholipases/chemistry , Phospholipases/genetics , Protein Structure, Tertiary , Retina/pathology , Zebrafish/embryology
10.
Nature ; 457(7229): 585-8, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19060881

ABSTRACT

Aeroallergy results from maladaptive immune responses to ubiquitous, otherwise innocuous environmental proteins. Although the proteins targeted by aeroallergic responses represent a tiny fraction of the airborne proteins humans are exposed to, allergenicity is a quite public phenomenon-the same proteins typically behave as aeroallergens across the human population. Why particular proteins tend to act as allergens in susceptible hosts is a fundamental mechanistic question that remains largely unanswered. The main house-dust-mite allergen, Der p 2, has structural homology with MD-2 (also known as LY96), the lipopolysaccharide (LPS)-binding component of the Toll-like receptor (TLR) 4 signalling complex. Here we show that Der p 2 also has functional homology, facilitating signalling through direct interactions with the TLR4 complex, and reconstituting LPS-driven TLR4 signalling in the absence of MD-2. Mirroring this, airway sensitization and challenge with Der p 2 led to experimental allergic asthma in wild type and MD-2-deficient, but not TLR4-deficient, mice. Our results indicate that Der p 2 tends to be targeted by adaptive immune responses because of its auto-adjuvant properties. The fact that other members of the MD-2-like lipid-binding family are allergens, and that most defined major allergens are thought to be lipid-binding proteins, suggests that intrinsic adjuvant activity by such proteins and their accompanying lipid cargo may have some generality as a mechanism underlying the phenomenon of allergenicity.


Subject(s)
Allergens/immunology , Allergens/metabolism , Antigens, Dermatophagoides/immunology , Antigens, Dermatophagoides/metabolism , Molecular Mimicry/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Air , Allergens/chemistry , Allergens/genetics , Animals , Antigens, Dermatophagoides/chemistry , Antigens, Dermatophagoides/genetics , Arthropod Proteins , Asthma/genetics , Asthma/immunology , Cell Line , Disease Models, Animal , Female , Humans , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/deficiency , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Mice , Protein Binding , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
11.
Proc Natl Acad Sci U S A ; 109(41): 16630-5, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23012424

ABSTRACT

Airway mucus plays a critical role in clearing inhaled toxins, particles, and pathogens. Diverse toxic, inflammatory, and infectious insults induce airway mucus secretion and goblet cell metaplasia to preserve airway sterility and homeostasis. However, goblet cell metaplasia, mucus hypersecretion, and airway obstruction are integral features of inflammatory lung diseases, including asthma, chronic obstructive lung disease, and cystic fibrosis, which cause an immense burden of morbidity and mortality. These chronic lung diseases are united by susceptibility to microbial colonization and recurrent airway infections. Whether these twinned phenomena (mucous metaplasia, compromised host defenses) are causally related has been unclear. Here, we demonstrate that SAM pointed domain ETS factor (SPDEF) was induced by rhinoviral infection of primary human airway cells and that cytoplasmic activities of SPDEF, a transcriptional regulator of airway goblet cell metaplasia, inhibited Toll-like receptor (TLR) activation of epithelial cells. SPDEF bound to and inhibited activities of TLR signaling adapters, MyD88 and TRIF, inhibiting MyD88-induced cytokine production and TRIF-induced interferon ß production. Conditional expression of SPDEF in airway epithelial cells in vivo inhibited LPS-induced neutrophilic infiltration and bacterial clearance. SPDEF-mediated inhibition of both TLR and type I interferon signaling likely protects the lung against inflammatory damage when inciting stimuli are not eradicated. Present findings provide, at least in part, a molecular explanation for increased susceptibility to infection in lung diseases associated with mucous metaplasia and a mechanism by which patients with florid mucous metaplasia may tolerate microbial burdens that are usually associated with fulminant inflammatory disease in normal hosts.


Subject(s)
Epithelial Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Blotting, Western , Doxycycline/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression/drug effects , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interleukin-13/pharmacology , Lipopolysaccharides/pharmacology , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lung Diseases/pathology , Metaplasia , Mice , Microscopy, Confocal , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Binding , Proto-Oncogene Proteins c-ets/genetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Rhinovirus/physiology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
12.
Cell Mol Life Sci ; 70(11): 1897-913, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22971774

ABSTRACT

The Eyes Absent (EYA) proteins, first described in the context of fly eye development, are now implicated in processes as disparate as organ development, innate immunity, DNA damage repair, photoperiodism, angiogenesis, and cancer metastasis. These functions are associated with an unusual combination of biochemical activities: tyrosine phosphatase and threonine phosphatase activities in separate domains, and transactivation potential when associated with a DNA-binding partner. EYA mutations are linked to multiorgan developmental disorders, as well as to adult diseases ranging from dilated cardiomyopathy to late-onset sensorineural hearing loss. With the growing understanding of EYA biochemical and cellular activity, biological function, and association with disease, comes the possibility that the EYA proteins are amenable to the design of targeted therapeutics. The availability of structural information, direct links to disease states, available animal models, and the fact that they utilize unconventional reaction mechanisms that could allow specificity, suggest that EYAs are well-positioned for drug discovery efforts. This review provides a summary of EYA structure, activity, and function, as they relate to development and disease, with particular emphasis on recent findings.


Subject(s)
Drosophila Proteins/physiology , Embryonic Development/genetics , Eye Proteins/physiology , Animals , Branchio-Oto-Renal Syndrome/genetics , Branchio-Oto-Renal Syndrome/pathology , Cell Movement/genetics , Cell Polarity/genetics , DNA Repair/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drosophila Proteins/chemistry , Eye Proteins/chemistry , Humans , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Physiologic/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/physiology , Trans-Activators/chemistry , Trans-Activators/metabolism , Trans-Activators/physiology
13.
Nat Commun ; 15(1): 1385, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360978

ABSTRACT

The Eyes Absent proteins (EYA1-4) are a biochemically unique group of tyrosine phosphatases known to be tumour-promoting across a range of cancer types. To date, the targets of EYA phosphatase activity remain largely uncharacterised. Here, we identify Polo-like kinase 1 (PLK1) as an interactor and phosphatase substrate of EYA4 and EYA1, with pY445 on PLK1 being the primary target site. Dephosphorylation of pY445 in the G2 phase of the cell cycle is required for centrosome maturation, PLK1 localization to centrosomes, and polo-box domain (PBD) dependent interactions between PLK1 and PLK1-activation complexes. Molecular dynamics simulations support the rationale that pY445 confers a structural impairment to PBD-substrate interactions that is relieved by EYA-mediated dephosphorylation. Depletion of EYA4 or EYA1, or chemical inhibition of EYA phosphatase activity, dramatically reduces PLK1 activation, causing mitotic defects and cell death. Overall, we have characterized a phosphotyrosine signalling network governing PLK1 and mitosis.


Subject(s)
Cell Cycle Proteins , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Tyrosine/metabolism , Mitosis , Centrosome/metabolism , Phosphoric Monoester Hydrolases/metabolism , HeLa Cells , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Trans-Activators/metabolism
14.
J Exp Clin Cancer Res ; 42(1): 273, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37858153

ABSTRACT

The insulin-like growth factor receptor (IGF-1R) was among the most intensively pursued kinase targets in oncology. However, even after a slew of small-molecule and antibody therapeutics reached clinical trials for a range of solid tumors, the initial promise remains unfulfilled. Mechanisms of resistance to, and toxicities resulting from, IGF-1R-targeted drugs are well-catalogued, and there is general appreciation of the fact that a lack of biomarker-based patient stratification was a limitation of previous clinical trials. But no next-generation therapeutic strategies have yet successfully exploited this understanding in the clinic.Currently there is emerging interest in re-visiting IGF-1R targeted therapeutics in combination-treatment protocols with predictive biomarker-driven patient-stratification. One such biomarker that emerged from early clinical trials is the sub-cellular localization of IGF-1R. After providing some background on IGF-1R, its drugging history, and the trials that led to the termination of drug development for this target, we look more deeply into the correlation between sub-cellular localization of IGF-1R and susceptibility to various classes of IGF-1R - targeted agents.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers
15.
Clin Cancer Res ; 29(2): 458-471, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36394520

ABSTRACT

PURPOSE: Targeted cancer therapeutics have not significantly benefited patients with Ewing sarcoma with metastatic or relapsed disease. Understanding the molecular underpinnings of drug resistance can lead to biomarker-driven treatment selection. EXPERIMENTAL DESIGN: Receptor tyrosine kinase (RTK) pathway activation was analyzed in tumor cells derived from a panel of Ewing sarcoma tumors, including primary and metastatic tumors from the same patient. Phospho-RTK arrays, Western blots, and IHC were used. Protein localization and the levels of key markers were determined using immunofluorescence. DNA damage tolerance was measured through PCNA ubiquitination levels and the DNA fiber assay. Effects of pharmacologic inhibition were assessed in vitro and key results validated in vivo using patient-derived xenografts. RESULTS: Ewing sarcoma tumors fell into two groups. In one, IGF1R was predominantly nuclear (nIGF1R), DNA damage tolerance pathway was upregulated, and cells had low replication stress and RRM2B levels and high levels of WEE1 and RAD21. These tumors were relatively insensitive to IGF1R inhibition. The second group had high replication stress and RRM2B, low levels of WEE1 and RAD21, membrane-associated IGF1R (mIGF1R) signaling, and sensitivity to IGF1R or WEE1-targeted inhibitors. Moreover, the matched primary and metastatic tumors differed in IGF1R localization, levels of replication stress, and inhibitor sensitivity. In all instances, combined IGF1R and WEE1 inhibition led to tumor regression. CONCLUSIONS: IGF1R signaling mechanisms and replication stress levels can vary among Ewing sarcoma tumors (including in the same patient), influencing the effects of IGF1R and WEE1 treatment. These findings make the case for using biopsy-derived predictive biomarkers at multiple stages of Ewing sarcoma disease management.


Subject(s)
Sarcoma, Ewing , Humans , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , DNA Damage , Cell Line, Tumor , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Cell Cycle Proteins , Receptor, IGF Type 1/metabolism
16.
Nucleic Acids Res ; 38(11): 3827-33, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20185566

ABSTRACT

We have investigated the anomalously weak binding of human papillomavirus (HPV) regulatory protein E2 to a DNA target containing the spacer sequence TATA. Experiments in magnesium (Mg(2+)) and calcium (Ca(2+)) ion buffers revealed a marked reduction in cutting by DNase I at the CpG sequence in the protein-binding site 3' to the TATA spacer sequence, Studies of the cation dependence of DNA-E2 affinities showed that upon E2 binding the TATA sequence releases approximately twice as many Mg(2+) ions as the average of the other spacer sequences. Binding experiments for TATA spacer relative to ATAT showed that in potassium ion (K(+)) the E2 affinity of the two sequences is nearly equal, but the relative dissociation constant (K(d)) for TATA increases in the order K(+ )< Na(+ )< Ca(2+ )< Mg(2+). Except for Mg(2+), K(d) for TATA relative to ATAT is independent of ion concentration, whereas for Mg(2+) the affinity for TATA drops sharply as ion concentration increases. Thus, ions of increasing positive charge density increasingly distort the E2 binding site, weakening the affinity for protein. In the case of Mg(2+), additional ions are bound to TATA that require displacement for protein binding. We suggest that the TATA sequence may bias the DNA structure towards a conformation that binds the protein relatively weakly.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/chemistry , Oncogene Proteins, Viral/metabolism , Base Sequence , DNA/metabolism , Deoxyribonuclease I/metabolism , Magnesium/chemistry , Nucleic Acid Conformation , Protein Binding
17.
BMC Med Genet ; 12: 21, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21306635

ABSTRACT

BACKGROUND: Recessive mutations of fibroblast growth factor 3 (FGF3) can cause LAMM syndrome (OMIM 610706), characterized by fully penetrant complete labyrinthine aplasia, microtia and microdontia. METHODS: We performed a prospective molecular genetic and clinical study of families segregating hearing loss linked to FGF3 mutations. Ten affected individuals from three large Pakistani families segregating FGF3 mutations were imaged with CT, MRI, or both to detect inner ear abnormalities. We also modeled the three dimensional structure of FGF3 to better understand the structural consequences of the three missense mutations. RESULTS: Two families segregated reported mutations (p.R104X and p.R95W) and one family segregated a novel mutation (p.R132GfsX26) of FGF3. All individuals homozygous for p.R104X or p.R132GfsX26 had fully penetrant features of LAMM syndrome. However, recessive p.R95W mutations were associated with nearly normal looking auricles and variable inner ear structural phenotypes, similar to that reported for a Somali family also segregating p.R95W. This suggests that the mild phenotype is not entirely due to genetic background. Molecular modeling result suggests a less drastic effect of p.R95W on FGF3 function compared with known missense mutations detected in fully penetrant LAMM syndrome. Since we detected significant intrafamilial variability of the inner ear structural phenotype in the family segregating p.R95W, we also sequenced FGF10 as a likely candidate for a modifier. However, we did not find any sequence variation, pointing out that a larger sample size will be needed to map and identify a modifier. We also observed a mild to moderate bilateral conductive hearing loss in three carriers of p.R95W, suggesting either a semi-dominant effect of this mutant allele of FGF3, otitis media, or a consequence of genetic background in these three family members. CONCLUSIONS: We noted a less prominent dental and external ear phenotype in association with the homozygous p.R95W. Therefore, we conclude that the manifestations of recessive FGF3 mutations range from fully penetrant LAMM syndrome to deafness with residual inner ear structures and, by extension, with minimal syndromic features, an observation with implications for cochlear implantation candidacy.


Subject(s)
Deafness/genetics , Fibroblast Growth Factor 3/genetics , Hearing Loss/genetics , Congenital Abnormalities/genetics , Congenital Microtia , DNA Mutational Analysis , Ear/abnormalities , Ear, Inner/abnormalities , Female , Gene Expression , Homozygote , Humans , Male , Mutation , Pedigree , Tooth Abnormalities
18.
Expert Rev Respir Med ; 15(12): 1539-1549, 2021 12.
Article in English | MEDLINE | ID: mdl-34758677

ABSTRACT

INTRODUCTION: Mycophenolate mofetil (MMF), initially approved to prevent rejection in solid organ allograft, is now being increasingly used for other conditions. Over the last decade, MMF has emerged as a useful therapy for a variety of immune-mediated diseases. AREAS COVERED: There has been a growing interest in the clinical use of MMF in the treatment of ILDs due to its versatile anti-inflammatory, immunomodulatory, anti-fibrotic and anti-proliferative properties. In this focussed review, we summarize the available literature using the Pubmed, Science Direct and EMBASE databases published until June 2021 on the efficacy and tolerability of MMF in various ILDs. EXPERT OPINION: Other than idiopathic pulmonary fibrosis (IPF) and its broader category of progressive fibrosing ILD, there have been no drugs approved by relevant regulatory agencies for the treatment of the multiple other forms of ILD. Though results are limited, immunosuppressants such as MMF have shown promise as an effective and well-tolerated steroid-sparing agent, providing hope that the limited treatment armamentarium for ILDs can be expanded.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , Immunosuppressive Agents/adverse effects , Lung , Lung Diseases, Interstitial/drug therapy , Mycophenolic Acid/adverse effects
19.
Mol Cancer Ther ; 20(5): 803-815, 2021 05.
Article in English | MEDLINE | ID: mdl-33649104

ABSTRACT

EWSR1/FLI1, the most common fusion gene in Ewing sarcoma, upregulates expression of the Eyes Absent 3 (EYA3) transactivator-phosphatase protein. The purpose of this study was to investigate molecular and cellular mechanisms through which EYA3 might promote Ewing sarcoma tumor growth and to determine whether the EYA3 tyrosine phosphatase activity represents a viable therapeutic target. We used genetic and pharmacologic modulation of EYA3 in cell line-based xenografts to examine how loss of EYA3 tyrosine phosphatase activity affects tumor growth and angiogenesis. Molecular mechanisms were evaluated in vivo and in vitro through analyses of tumor tissue and multicellular tumor spheroids. Our results show that both loss of EYA3 in Ewing sarcoma cells and pharmacologic inhibition of the EYA3 tyrosine phosphatase activity inhibit tumor growth and tumor angiogenesis. EYA3 regulates levels of VEGFA in Ewing tumors, as well as promoting DNA damage repair and survival of Ewing sarcoma tumor cells. Target engagement is demonstrated in tumor tissue through elevated levels of the EYA3 substrate H2AX-pY142 upon loss of EYA3 or with Benzarone treatment. The efficacy of EYA3 tyrosine phosphatase inhibition in attenuating tumor growth and angiogenesis is corroborated in an Ewing sarcoma patient-derived tumor xenograft. Together, the results presented here validate EYA3 as a target for the development of novel Ewing sarcoma therapeutic strategies, and set the stage for evaluating the efficacy of combining the antiangiogenic and anti-cell survival effects of EYA3 inhibition with cytotoxic chemotherapy.


Subject(s)
DNA Damage/genetics , DNA-Binding Proteins/metabolism , Sarcoma, Ewing/therapy , Animals , Cell Line, Tumor , Female , Humans , Mice , Neovascularization, Pathologic , Sarcoma, Ewing/pathology
20.
Nature ; 426(6964): 295-8, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14628052

ABSTRACT

The Eyes absent proteins are members of a conserved regulatory network implicated in the development of the eye, muscle, kidney and ear. Mutations in the Eyes absent genes have been associated with several congenital disorders including the multi-organ disease bronchio-oto-renal syndrome, congenital cataracts and late-onset deafness. On the basis of previous analyses it has been shown that Eyes absent is a nuclear transcription factor, acting through interaction with homeodomain-containing Sine oculis (also known as Six) proteins. Here we show that Eyes absent is also a protein tyrosine phosphatase. It does not resemble the classical tyrosine phosphatases that use cysteine as a nucleophile and proceed by means of a thiol-phosphate intermediate. Rather, Eyes absent is the prototype for a class of protein tyrosine phosphatases that use a nucleophilic aspartic acid in a metal-dependent reaction. Furthermore, the phosphatase activity of Eyes absent contributes to its ability to induce eye formation in Drosophila.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Eye Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Animals , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/chemistry , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Eye/embryology , Eye/enzymology , Eye/metabolism , Eye Proteins/antagonists & inhibitors , Eye Proteins/chemistry , Molecular Sequence Data , Mutation , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL