Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
EMBO J ; 43(7): 1325-1350, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38321267

ABSTRACT

Exit from mitosis is brought about by dramatic changes in the phosphoproteome landscape. A drop in Cyclin-dependent kinase (Cdk) activity, the master regulatory kinase, and activation of counteracting phosphatases such as Cdc14 in budding yeast, results in ordered substrate dephosphorylation, allowing entry into a new cell cycle and replication licensing. In meiosis however, two cell divisions have to be executed without intermediate DNA replication, implying that global phosphorylation and dephosphorylation have to be adapted to the challenges of meiosis. Using a global time-resolved phosphoproteomics approach in budding yeast, we compared the phosphoproteome landscape between mitotic exit and the transition from meiosis I to meiosis II. We found that unlike exit from mitosis, Cdk phosphomotifs remain mostly stably phosphorylated at the end of meiosis I, whereas a majority of Cdk-unrelated motifs are reset by dephosphorylation. However, inducing an artificial drop of Cdk at metaphase of meiosis I leads to ordered substrate dephosphorylation, comparable to mitosis, indicating that phosphoregulation of substrates at the end of meiosis I is thus mainly qualitatively rather than quantitatively ordered.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomycetales , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Saccharomycetales/genetics , Saccharomycetales/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Mitosis , Phosphorylation , Meiosis
2.
J Biol Chem ; 300(7): 107408, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796066

ABSTRACT

The eyes absent (Eya) proteins were first identified as co-activators of the six homeobox family of transcription factors and are critical in embryonic development. These proteins are also re-expressed in cancers after development is complete, where they drive tumor progression. We have previously shown that the Eya3 N-terminal domain (NTD) contains Ser/Thr phosphatase activity through an interaction with the protein phosphatase 2A (PP2A)-B55α holoenzyme and that this interaction increases the half-life of Myc through pT58 dephosphorylation. Here, we showed that Eya3 directly interacted with the NTD of Myc, recruiting PP2A-B55α to Myc. We also showed that Eya3 increased the Ser/Thr phosphatase activity of PP2A-B55α but not PP2A-B56α. Furthermore, we demonstrated that the NTD (∼250 amino acids) of Eya3 was completely disordered, and it used a 38-residue segment to interact with B55α. In addition, knockdown and phosphoproteomic analyses demonstrated that Eya3 and B55α affected highly similar phosphosite motifs with a preference for Ser/Thr followed by Pro, consistent with Eya3's apparent Ser/Thr phosphatase activity being mediated through its interaction with PP2A-B55α. Intriguingly, mutating this Pro to other amino acids in a Myc peptide dramatically increased dephosphorylation by PP2A. Not surprisingly, MycP59A, a naturally occurring mutation hotspot in several cancers, enhanced Eya3-PP2A-B55α-mediated dephosphorylation of pT58 on Myc, leading to increased Myc stability and cell proliferation, underscoring the critical role of this phosphosite in regulating Myc stability.


Subject(s)
Protein Phosphatase 2 , Proto-Oncogene Proteins c-myc , Humans , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Protein Binding , HEK293 Cells , Protein Domains , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/chemistry , DNA-Binding Proteins
3.
J Biomed Sci ; 31(1): 33, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38532423

ABSTRACT

BACKGROUND: T cell receptor (TCR) signaling and T cell activation are tightly regulated by gatekeepers to maintain immune tolerance and avoid autoimmunity. The TRAIL receptor (TRAIL-R) is a TNF-family death receptor that transduces apoptotic signals to induce cell death. Recent studies have indicated that TRAIL-R regulates T cell-mediated immune responses by directly inhibiting T cell activation without inducing apoptosis; however, the distinct signaling pathway that regulates T cell activation remains unclear. In this study, we screened for intracellular TRAIL-R-binding proteins within T cells to explore the novel signaling pathway transduced by TRAIL-R that directly inhibits T cell activation. METHODS: Whole-transcriptome RNA sequencing was used to identify gene expression signatures associated with TRAIL-R signaling during T cell activation. High-throughput screening with mass spectrometry was used to identify the novel TRAIL-R binding proteins within T cells. Co-immunoprecipitation, lipid raft isolation, and confocal microscopic analyses were conducted to verify the association between TRAIL-R and the identified binding proteins within T cells. RESULTS: TRAIL engagement downregulated gene signatures in TCR signaling pathways and profoundly suppressed phosphorylation of TCR proximal tyrosine kinases without inducing cell death. The tyrosine phosphatase SHP-1 was identified as the major TRAIL-R binding protein within T cells, using high throughput mass spectrometry-based proteomics analysis. Furthermore, Lck was co-immunoprecipitated with the TRAIL-R/SHP-1 complex in the activated T cells. TRAIL engagement profoundly inhibited phosphorylation of Lck (Y394) and suppressed the recruitment of Lck into lipid rafts in the activated T cells, leading to the interruption of proximal TCR signaling and subsequent T cell activation. CONCLUSIONS: TRAIL-R associates with phosphatase SHP-1 and transduces a unique and distinct immune gatekeeper signal to repress TCR signaling and T cell activation via inactivating Lck. Thus, our results define TRAIL-R as a new class of immune checkpoint receptors for restraining T cell activation, and TRAIL-R/SHP-1 axis can serve as a potential therapeutic target for immune-mediated diseases.


Subject(s)
Receptors, Antigen, T-Cell , Receptors, TNF-Related Apoptosis-Inducing Ligand , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Antigen, T-Cell/metabolism , Jurkat Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Phosphorylation , Lymphocyte Activation , Tyrosine/metabolism
4.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125727

ABSTRACT

Branchio-oto-renal (BOR) and branchio-otic (BO) syndromes are characterized by anomalies affecting the ears, often accompanied by hearing loss, as well as abnormalities in the branchial arches and renal system. These syndromes exhibit a broad spectrum of phenotypes and a complex genomic landscape, with significant contributions from the EYA1 gene and the SIX gene family, including SIX1 and SIX5. Due to their diverse phenotypic presentations, which can overlap with other genetic syndromes, molecular genetic confirmation is essential. As sequencing technologies advance, whole-genome sequencing (WGS) is increasingly used in rare disease diagnostics. We explored the genomic landscape of 23 unrelated Korean families with typical or atypical BOR/BO syndrome using a stepwise approach: targeted panel sequencing and exome sequencing (Step 1), multiplex ligation-dependent probe amplification (MLPA) with copy number variation screening (Step 2), and WGS (Step 3). Integrating WGS into our diagnostic pipeline detected structure variations, including cryptic inversion and complex genomic rearrangement, eventually enhancing the diagnostic yield to 91%. Our findings expand the genomic architecture of BOR/BO syndrome and highlight the need for WGS to address the genetic diagnosis of clinically heterogeneous rare diseases.


Subject(s)
Branchio-Oto-Renal Syndrome , DNA Copy Number Variations , Whole Genome Sequencing , Humans , Branchio-Oto-Renal Syndrome/genetics , Republic of Korea , Whole Genome Sequencing/methods , Female , Male , DNA Copy Number Variations/genetics , Intracellular Signaling Peptides and Proteins/genetics , Rare Diseases/genetics , Nuclear Proteins/genetics , Homeodomain Proteins/genetics , Child , Protein Tyrosine Phosphatases/genetics , Child, Preschool , Adult , Genomics/methods , Phenotype , Pedigree , Adolescent , Infant
5.
Methods Mol Biol ; 2743: 123-133, 2024.
Article in English | MEDLINE | ID: mdl-38147212

ABSTRACT

Protein tyrosine phosphorylation and dephosphorylation are key regulatory mechanisms in eukaryotes. Protein tyrosine phosphorylation and dephosphorylation are catalyzed by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), respectively. The combinatorial action of both PTKs and PTPs is essential for properly maintaining cellular functions. In this unit, we discuss different novel methods to identify PTP substrates. PTPs depend on specific invariant residues that enable binding to tyrosine-phosphorylated substrates and aid catalytic activity. Identifying PTP substrates has paved the way to understanding their role in distinct intracellular signaling pathways. Due to their high specific activity, the interaction between PTPs and their substrates is transient; therefore, identifying the physiological substrates of PTPs has been challenging. To identify the physiological substrates of PTPs, various PTP mutants have been generated. These PTP mutants, named "substrate-trapping mutants," lack catalytic activity but bind tightly to their tyrosine-phosphorylated substrates. Identifying the substrates for the PTPs will provide critical insight into the function of physiological and pathophysiological signal transduction. In this chapter, we describe interaction assays used to identify the PTP substrates.


Subject(s)
Protein Tyrosine Phosphatases , Signal Transduction , Protein Tyrosine Phosphatases/genetics , Phosphorylation , Protein-Tyrosine Kinases , Tyrosine
6.
Methods Mol Biol ; 2743: 271-283, 2024.
Article in English | MEDLINE | ID: mdl-38147221

ABSTRACT

Protein tyrosine phosphatases (PTPs) are important therapeutic targets for a range of human pathologies. However, the common architecture of PTP active sites impedes the discovery of selective PTP inhibitors. Our laboratory has recently developed methods to inhibit PTPs allosterically by targeting cysteine residues that either (i) are not conserved in the PTP family or (ii) result from pathogenic mutations. Here, we describe screening protocols for the identification of selective inhibitors that covalently engage such "rare" cysteines in target PTPs. Moreover, to elucidate the breadth of possible applications of our cysteine-directed screening protocols, we provide a brief overview of the nonconserved cysteines present in all human classical PTP domains.


Subject(s)
Cysteine , Protein Tyrosine Phosphatases , Humans , Mutation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics
7.
Methods Mol Biol ; 2743: 153-163, 2024.
Article in English | MEDLINE | ID: mdl-38147214

ABSTRACT

Tyrosine phosphorylation regulates signaling network activity downstream of receptor tyrosine kinase (RTK) activation. Receptor protein tyrosine phosphatases (RPTPs) serve to dephosphorylate RTKs and their proximal adaptor proteins, thus serving to modulate RTK activity. While the general function of RPTPs is well understood, the direct and indirect substrates for each RPTP are poorly characterized. Here we describe a method, quantitative phosphotyrosine phosphoproteomics, that enables the identification of specific phosphorylation sites whose phosphorylation levels are altered by the expression and activity of a given RPTP. In a proof-of-concept application, we use this method to highlight several direct or indirect substrate phosphorylation sites for PTPRJ, also known as DEP1, and show their quantitative phosphorylation in the context of wild-type PTPRJ compared to a mutant form of PTPRJ with increased activity, in EGF-stimulated cells. This method is generally applicable to define the signaling network effects of each RPTP in cells or tissues under different physiological conditions.


Subject(s)
Protein Tyrosine Phosphatases , Signal Transduction , Protein Tyrosine Phosphatases/genetics , Adaptor Proteins, Signal Transducing , Phosphorylation , Protein Processing, Post-Translational
8.
Methods Mol Biol ; 2743: 1-19, 2024.
Article in English | MEDLINE | ID: mdl-38147205

ABSTRACT

Nonsense mutations generating premature termination codons (PTCs) in various genes are frequently associated with somatic cancer and hereditary human diseases since PTCs commonly generate truncated proteins with defective or altered function. Induced translational readthrough during protein biosynthesis facilitates the incorporation of an amino acid at the position of a PTC, allowing the synthesis of a complete protein. This may evade the pathological effect of the PTC mutation and provide new therapeutic opportunities. Several protein tyrosine phosphatases (PTPs) genes are targeted by PTC in human disease, the tumor suppressor PTEN being the more prominent paradigm. Here, using PTEN and laforin as examples, two PTPs from the dual-specificity phosphatase subfamily, we describe methodologies to analyze in silico the distribution and frequency of pathogenic PTC in PTP genes. We also summarize laboratory protocols and technical notes to study the induced translational readthrough reconstitution of the synthesis of PTP targeted by PTC in association with disease in cellular models.


Subject(s)
Codon, Nonsense , Protein Tyrosine Phosphatases , Humans , Mutation , Protein Tyrosine Phosphatases/genetics , Dual-Specificity Phosphatases , Protein Biosynthesis
9.
Methods Mol Biol ; 2743: 93-110, 2024.
Article in English | MEDLINE | ID: mdl-38147210

ABSTRACT

The zebrafish is an ideal model for functional analysis of genes at the molecular, protein, cell, organ, and organism levels. We have used zebrafish to analyze the function of members of the protein tyrosine phosphatase (PTP) superfamily for more than two decades. The molecular genetic toolbox has significantly improved over the years. Currently, generating mutant lines that lack the function of a PTP gene is relatively straightforward by CRISPR/Cas9 technology-mediated generation of insertions or deletions in the target gene. In addition, generating point mutations using CRISPR/Cas9 technology and homology-directed repair (HDR) is feasible, albeit the success rate could be higher. Here, we describe the methods, including the tips and tricks, that we have used to generate knock-out and knock-in zebrafish lines in PTP genes successfully.


Subject(s)
Craniocerebral Trauma , Perciformes , Animals , Zebrafish/genetics , Protein Tyrosine Phosphatases/genetics , Point Mutation
10.
Methods Mol Biol ; 2743: 195-209, 2024.
Article in English | MEDLINE | ID: mdl-38147217

ABSTRACT

Receptor protein tyrosine phosphatases (RPTPs) are one of the key regulators of receptor tyrosine kinases (RTKs) and therefore play a critical role in modulating signal transduction. While the structure-function relationship of RTKs has been widely studied, the mechanisms modulating the activity of RPTPs still need to be fully understood. On the other hand, homodimerization has been shown to antagonize RPTP catalytic activity and appears to be a general feature of the entire family. Conversely, their documented ability to physically interact with RTKs is integral to their negative regulation of RTKs, but there is a yet-to-be proposed common model. However, specific transmembrane (TM) domain interactions and residues have been shown to be essential in regulating RPTP homodimerization, interactions with RTK substrates, and activity. Therefore, elucidating the contribution of the TM domains in RPTP regulation can provide significant insights into how these receptors function, interact, and eventually be modulated. This chapter describes the dominant-negative AraC-based transcriptional reporter (DN-AraTM) assay to identify specific TM interactions essential to homodimerization and heteroassociation with other membrane receptors, such as RTKs.


Subject(s)
Protein Tyrosine Phosphatases , Signal Transduction , Protein Tyrosine Phosphatases/genetics , Biological Assay , Protein Domains , Receptor Protein-Tyrosine Kinases
11.
BMC Med Genomics ; 17(1): 89, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627775

ABSTRACT

OBJECTIVE: Branchio-oto-renal syndrome (BOR, OMIM#113,650) is a rare autosomal dominant disorder that presents with a variety of symptoms, including hearing loss (sensorineural, conductive, or mixed), structural abnormalities affecting the outer, middle, and inner ear, branchial fistulas or cysts, as well as renal abnormalities.This study aims to identify the pathogenic variants by performing genetic testing on a family with Branchio-oto-renal /Branchio-otic (BO, OMIM#602,588) syndrome using whole-exome sequencing, and to explore possible pathogenic mechanisms. METHODS: The family spans 4 generations and consists of 9 individuals, including 4 affected by the BOR/BO syndrome. Phenotypic information, including ear malformation and branchial cleft, was collected from family members. Audiological, temporal bone imaging, and renal ultrasound examinations were also performed. Whole-exome sequencing was conducted to identify candidate pathogenic variants and explore the underlying molecular etiology of BOR/BO syndrome by minigene experiments. RESULTS: Intra-familial variability was observed in the clinical phenotypes of BOR/BO syndrome in this family. The severity and nature of hearing loss varied in family members, with mixed or sensorineural hearing loss. The proband, in particular, had profound sensorineural hearing loss on the left and moderate conductive hearing loss on the right. Additionally, the proband exhibited developmental delay, and her mother experienced renal failure during pregnancy and terminated the pregnancy prematurely. Genetic testing revealed a novel heterozygous variant NM_000503.6: c.639 + 3 A > C in the EYA1 gene in affected family members. In vitro minigene experiments demonstrated its effect on splicing. According to the American College of Medical Genetics (ACMG) guidelines, this variant was classified as likely pathogenic. CONCLUSION: This study highlights the phenotypic heterogeneity within the same family, reports the occurrence of renal failure and adverse pregnancy outcomes in a female patient at reproductive age with BOR syndrome, and enriches the mutational spectrum of pathogenic variants in the EYA1 gene.


Subject(s)
Branchio-Oto-Renal Syndrome , Deafness , Hearing Loss, Sensorineural , Hearing Loss , Renal Insufficiency , Humans , Pregnancy , Female , Branchio-Oto-Renal Syndrome/genetics , Branchio-Oto-Renal Syndrome/pathology , Intracellular Signaling Peptides and Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Hearing Loss/genetics , Pedigree , Nuclear Proteins/genetics
12.
Open Biol ; 14(2): 230278, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38378139

ABSTRACT

Neuroparasitism concerns the hostile take-over of a host's nervous system by a foreign invader, in order to alter the behaviour of the host in favour of the parasite. One of the most remarkable cases of parasite-induced host behavioural manipulation comprises the changes baculoviruses induce in their caterpillar hosts. Baculoviruses may manipulate caterpillar behaviour in two ways: hyperactivity (increased movement in the horizontal plane) and/or tree-top disease (movement to elevated levels in the vertical plane). Those behavioural changes are followed by liquefaction and death of the caterpillar. In Autographa californica multiple nucleopolyhedrovirus (AcMNPV)-infected Spodoptera exigua caterpillars, an enzymatic active form of the virally encoded protein tyrosine phosphatase (PTP) is needed for the expression of hyperactivity from 3 days post infection (dpi). Using eGFP-expressing recombinant AcMNPV strains, we show that infection of the caterpillar's central nervous system (CNS) can be observed primarily from 3 dpi onwards. In addition, we demonstrate that the structural and enzymatic function of PTP does not play a role in infection of the CNS. Instead we show that the virus entered the CNS via the trachea, progressing caudally to frontally through the CNS and that the infection progressed from the outermost cell layers towards the inner cell layers of the CNS, in a PTP independent manner. These findings help to further understand parasitic manipulation and the mechanisms by which neuroparasites infect the host nervous system to manipulate host behaviour.


Subject(s)
Baculoviridae , Central Nervous System , Nucleopolyhedroviruses , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Spodoptera/metabolism , Central Nervous System/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism
13.
Article in Zh | MEDLINE | ID: mdl-38973045

ABSTRACT

Objective:To investigate the clinical phenotype of a family with branchio-oto syndrome (BOS) and to explore the genetic etiology of the syndrome in this family. Methods:Clinical data were collected from a child diagnosed with BOS and his family members. Genomic DNA was extracted from peripheral blood of the proband and his family members. Whole-exome sequencing was performed, and the mutation sites were verified and analyzed by Sanger sequencing. Results:The family consists of two generations with four members, three of whom exhibit the phenotype. Two members have hearing loss and bilateral preauricular fistulas and bilateral branchial cleft fistulas. One member has bilateral preauricular fistulas and bilateral branchial cleft fistulas. All of which were in line with the clinical diagnosis of gill ear syndrome, the inheritance mode of the family was autosomal dominant inheritance, genetic testing showed that all members of the family had c. 1744delC(p. L592Cfs*47) mutation in the EYA1 gene, while unaffected members have the wild-type allele at this locus. This mutation is a frameshift mutation, which results in the early appearance of the stop codon, and has not been reported so far. According to ACMG guidelines, the variant was preliminarily determined to be suspected pathogenic. Conclusion:The newly discovered EYA1c. 1744delC(p. L592Cfs*47) mutation in this family is the pathogenic mutant gene of the patients in this family, which further expands the mutation spectrum of EYA1 gene, gives us a new understanding of the disease, and provides an important reference for clinical diagnosis and genetic counseling.


Subject(s)
Intracellular Signaling Peptides and Proteins , Nuclear Proteins , Pedigree , Phenotype , Protein Tyrosine Phosphatases , Humans , Male , Protein Tyrosine Phosphatases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Female , Exome Sequencing , Branchio-Oto-Renal Syndrome/genetics , Frameshift Mutation , Mutation , Genetic Testing , Child , Adult
14.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189098, 2024 May.
Article in English | MEDLINE | ID: mdl-38555001

ABSTRACT

The Eya family of proteins (consisting of Eyas1-4 in mammals) play vital roles in embryogenesis by regulating processes such as proliferation, migration/invasion, cellular survival and pluripotency/plasticity of epithelial and mesenchymal states. Eya proteins carry out such diverse functions through a unique combination of transcriptional co-factor, Tyr phosphatase, and PP2A/B55α-mediated Ser/Thr phosphatase activities. Since their initial discovery, re-expression of Eyas has been observed in numerous tumor types, where they are known to promote tumor progression through a combination of their transcriptional and enzymatic activities. Eya proteins thus reinstate developmental processes during malignancy and represent a compelling class of therapeutic targets for inhibiting tumor progression.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatases , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/genetics , Animals , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
15.
DNA Repair (Amst) ; 141: 103729, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089192

ABSTRACT

The Eyes Absent family (EYA1-4) are a group of dual function proteins that act as both tyrosine phosphatases and transcriptional co-activators. EYA proteins play a vital role in development, but are also aberrantly overexpressed in cancers, where they often confer an oncogenic effect. Precisely how the EYAs impact cell biology is of growing interest, fuelled by the therapeutic potential of an expanding repertoire of EYA inhibitors. Recent functional studies suggest that the EYAs are important players in the regulation of genome maintenance pathways including DNA repair, mitosis, and DNA replication. While the characterized molecular mechanisms have predominantly been ascribed to EYA phosphatase activities, EYA co-transcriptional activity has also been found to impact the expression of genes that support these pathways. This indicates functional convergence of EYA phosphatase and co-transcriptional activities, highlighting the emerging importance of the EYA protein family at the intersection of genome maintenance mechanisms. In this review, we discuss recent progress in defining EYA protein substrates and transcriptional effects, specifically in the context of genome maintenance. We then outline future directions relevant to the field and discuss the clinical utility of EYA inhibitors.


Subject(s)
DNA Repair , DNA Replication , Mitosis , Protein Tyrosine Phosphatases , Humans , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/genetics , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism
16.
Mem. Inst. Oswaldo Cruz ; 107(6): 713-719, set. 2012. ilus, tab
Article in English | LILACS | ID: lil-649484

ABSTRACT

Protein tyrosine phosphatases (PTPs) play an essential role in the regulation of cell differentiation in pathogenic trypanosomatids. In this study, we describe a PTP expressed by the non-pathogenic protozoan Trypanosoma rangeli (TrPTP2). The gene for this PTP is orthologous to the T. brucei TbPTP1 and Trypanosoma cruzi (TcPTP2) genes. Cloning and expression of the TrPTP2 and TcPTP2 proteins allowed anti-PTP2 monoclonal antibodies to be generated in BALB/c mice. When expressed by T. rangeli epimastigotes and trypomastigotes, native TrPTP2 is detected as a ~65 kDa protein associated with the parasite's flagellum. Given that the flagellum is an important structure for cell differentiation in trypanosomatids, the presence of a protein responsible for tyrosine dephosphorylation in the T. rangeli flagellum could represent an interesting mechanism of regulation in this structure.


Subject(s)
Animals , Mice , Antibodies, Monoclonal/immunology , Flagella/enzymology , Protein Tyrosine Phosphatases/metabolism , Trypanosoma rangeli/enzymology , Immunization , Mice, Inbred BALB C , Phylogeny , Protein Tyrosine Phosphatases/genetics , Trypanosoma rangeli/genetics , Trypanosoma rangeli/immunology
17.
Santafé de Bogotá; s.n; 2000. 112 p. ilus, tab, graf.
Thesis in Spanish | LILACS | ID: lil-278179

ABSTRACT

El cáncer es una enfermedad del genoma, en el cual múltiples aberraciones genéticas conducen a la activación de oncogenes y a la inactivación de genes supresores tumorales, promoviendo un desequilibrio en el control de la proliferación, diferenciación y muertecelular. Las células tumorales pueden ser inducidas a un crecimiento controlado y a diferenciación terminal in vitro por tratamiento farmacológico que conduce a la restauración parcial del fenotipo celular normal. Sin embargo, el uso de inductores farmacológicos de diferenciarón, supresión del crecimiento y muerte celular como el ácido retinoíco, vitamina D, L-tirosina y bromodeoxiuridina, es limitado debido a que no se conocen los mecanismos moleculares y sus blancos de acción. La Bromodeoxiuridina, es un análogo de la timina, sensibilizador a la radiación ultravioleta e inductor de la supresión del crecimiento. Sin embargo, las bases moleculares de estas acciones no se conocen muy bien. La identificación de genes blancos moleculares (como cinasas y fosfatasas) de la Bromodeoxiuridina (BrdU) y su papel en el control de la proliferación celular y la radiosensibilización, conduciría al entendimiento y desarrollo de potenciales agentes terapéuticos para el control de la proliferación en células tumorales, como tratamiento alternativo a la quimioterapia y radioterapia convencionales, las cuales son altamente agresivas no sólo para las células tumorales, sino también para las células normales en proliferación. Los genes que codifican para proteínas fosfatasas se han implicado en la tumorigénesis. Recientemente en el laboratorio de fisiología molecular del INS se clonó y se secuenció el cADN que codifica para la tirosina fosfatasa PRL-1. Sin embargo, no se sabe si sus niveles de expresión cambian en células de melanoma proliferantes e inducidas a supresión del crecimiento y diferenciación celular. Por lo tanto, el objetivo de este trabajo fue determinar el nivel de expresión de ARN del gen que codifica para la tirosina-fosfatasa PRL-1, en células de melanoma murino B-16 inducidas a supresión del crecimiento y diferenciación celular. Por lo tanto, el objetivo de este trabajo fue determinar el nivel de expresión del ARN del gen que codifica para la tirosina-fosfatasa PRL-1, en células de melanoma murino B-16 inducidas a supresión del crecimiento por el tratamiento con Bromodeoxiuridina, radiación ultravioleta tipo C y sensibilización...


Subject(s)
Bromodeoxyuridine/therapeutic use , Academic Dissertations as Topic , Gene Expression/radiation effects , In Vitro Techniques , Melanoma, Experimental/radiotherapy , Protein Tyrosine Phosphatases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL