Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Mol Biol Cell ; 32(22): ar42, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34586919

RESUMEN

The family of Bro1 proteins coordinates the activity of the Endosomal Sorting Complexes Required for Transport (ESCRTs) to mediate a number of membrane remodeling events. These events culminate in membrane scission catalyzed by ESCRT-III, whose polymerization and disassembly is controlled by the AAA-ATPase, Vps4. Bro1-family members Alix and HD-PTP as well as yeast Bro1 have central "V" domains that noncovalently bind Ub and connect ubiquitinated proteins to ESCRT-driven functions such as the incorporation of ubiquitinated membrane proteins into intralumenal vesicles of multivesicular bodies. Recently, it was discovered that the V domain of yeast Bro1 binds the MIT domain of Vps4 to stimulate its ATPase activity. Here we determine the structural basis for how the V domain of human HD-PTP binds ubiquitin. The HD-PTP V domain also binds the MIT domain of Vps4, and ubiquitin binding to the HD-PTP V domain enhances its ability to stimulate Vps4 ATPase activity. Additionally, we found that V domains of both HD-PTP and Bro1 bind CHMP5 and Vps60, respectively, providing another potential molecular mechanism to alter Vps4 activity. These data support a model whereby contacts between ubiquitin, ESCRT-III, and Vps4 by V domains of the Bro1 family may coordinate late events in ESCRT-driven membrane remodeling events.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Ubiquitina/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Sitios de Unión , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Humanos , Modelos Moleculares , Dominios y Motivos de Interacción de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética , Dispersión del Ángulo Pequeño , Técnicas del Sistema de Dos Híbridos , ATPasas de Translocación de Protón Vacuolares/genética , Difracción de Rayos X
2.
Clin Biochem ; 93: 80-89, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33831386

RESUMEN

OBJECTIVES: Mutations in the gene encoding the glycogen phosphatase laforin result in the fatal childhood dementia Lafora disease (LD). A cellular hallmark of LD is cytoplasmic, hyper-phosphorylated, glycogen-like aggregates called Lafora bodies (LBs) that form in nearly all tissues and drive disease progression. Additional tools are needed to define the cellular function of laforin, understand the pathological role of laforin in LD, and determine the role of glycogen phosphate in glycogen metabolism. In this work, we present the generation and characterization of laforin nanobodies, with one being a laforin inhibitor. DESIGN AND METHODS: We identify multiple classes of specific laforin-binding nanobodies and determine their binding epitopes using hydrogen deuterium exchange (HDX) mass spectrometry. Using para-nitrophenyl phosphate (pNPP) and a malachite gold-based assay specific for glucan phosphatase activity, we assess the inhibitory effect of one nanobody on laforin's catalytic activity. RESULTS: Six families of laforin nanobodies are characterized and their epitopes mapped. One nanobody is identified and characterized that serves as an inhibitor of laforin's phosphatase activity. CONCLUSIONS: The six generated and characterized laforin nanobodies, with one being a laforin inhibitor, are an important set of tools that open new avenues to define unresolved glycogen metabolism questions.


Asunto(s)
Inhibidores Enzimáticos/química , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas no Receptoras/química , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Animales , Bioensayo , Camélidos del Nuevo Mundo , Cromatografía en Gel , Inhibidores Enzimáticos/farmacología , Mapeo Epitopo , Glucógeno/metabolismo , Oro/química , Humanos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Enfermedad de Lafora/enzimología , Modelos Moleculares , Nitrofenoles/química , Compuestos Organometálicos/química , Compuestos Organofosforados/química , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Anticuerpos de Dominio Único/aislamiento & purificación
3.
J Mol Recognit ; 34(7): e2890, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33620127

RESUMEN

The specific regulation of PTPN18 protein to three HER2 phospho-peptides has been studied by molecular dynamics simulations and free energy calculations. The results revealed that the three HER2 phospho-peptides binding to the PTPN18 catalytic domain is energetically favorable due to substrate specificity of PTPN18, and moreover, the PTPN18 protein have significantly higher affinity to pY1248 peptide (-45.22 kcal/mol) than that of pY1112 (-25.3 kcal/mol) and pY1196 (-31.86 kcal/mol) peptides. Further, the binding of HER2 phospho-peptides to PTPN18 have also caused the closure of WPD-loop with the decrease of the centroid distances between the P-loop and the WPD loop. The WPD-loop closure of PTPN18 relates directly to the new hydrogen bond and hydrophobic interaction formations between the residues Tyr62, Asp64, Val65, Ala231, Arg235, and Ala273 in PTPN18 and Tyr(PO3) in the HER2 phospho-peptides, which suggests that these key residues would contribute to the specific regulation of PTPN18 to the substrates. The correlation analysis revealed the allosteric communication networks from the pY binding loop to the WPD loop through the structural change and the residue interactions in PTPN18. These results will be helpful to understand the specific regulation through the allosteric communication network in the PTPN18 catalytic domain.


Asunto(s)
Simulación de Dinámica Molecular , Fosfopéptidos/química , Proteínas Tirosina Fosfatasas no Receptoras/química , Receptor ErbB-2/química , Dominio Catalítico , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Fosfopéptidos/metabolismo , Unión Proteica , Conformación Proteica , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptor ErbB-2/metabolismo , Especificidad por Sustrato
4.
Mol Cells ; 44(1): 26-37, 2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-33431714

RESUMEN

Human papillomaviruses (HPVs) cause cellular hyperproliferation-associated abnormalities including cervical cancer. The HPV genome encodes two major viral oncoproteins, E6 and E7, which recruit various host proteins by direct interaction for proteasomal degradation. Recently, we reported the structure of HPV18 E7 conserved region 3 (CR3) bound to the protein tyrosine phosphatase (PTP) domain of PTPN14, a well-defined tumor suppressor, and found that this intermolecular interaction plays a key role in E7-driven transformation and tumorigenesis. In this study, we carried out a molecular analysis of the interaction between CR3 of HPV18 E7 and the PTP domain of PTPN21, a PTP protein that shares high sequence homology with PTPN14 but is putatively oncogenic rather than tumor-suppressive. Through the combined use of biochemical tools, we verified that HPV18 E7 and PTPN21 form a 2:2 complex, with a dissociation constant of 5 nM and a nearly identical binding manner with the HPV18 E7 and PTPN14 complex. Nevertheless, despite the structural similarities, the biological consequences of the E7 interaction were found to differ between the two PTP proteins. Unlike PTPN14, PTPN21 did not appear to be subjected to proteasomal degradation in HPV18-positive HeLa cervical cancer cells. Moreover, knockdown of PTPN21 led to retardation of the migration/invasion of HeLa cells and HPV18 E7-expressing HaCaT keratinocytes, which reflects its protumor activity. In conclusion, the associations of the viral oncoprotein E7 with PTPN14 and PTPN21 are similar at the molecular level but play different physiological roles.


Asunto(s)
Alphapapillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Genotipo , Humanos , Modelos Moleculares , Invasividad Neoplásica , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteolisis
5.
Eur J Med Chem ; 190: 112131, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32078861

RESUMEN

Cryptic pockets, which are not apparent in crystallographic structures, provide promising alternatives to traditional binding sites for drug development. However, identifying cryptic pockets is extremely challenging and the therapeutic potential of cryptic pockets remains unclear. Here, we reported the discovery of novel inhibitors for striatal-enriched protein tyrosine phosphatase (STEP), a potential drug target for multiple neuropsychiatric disorders, based on cryptic pocket detection. By combining the use of molecular dynamics simulations and fragment-centric topographical mapping, we identified transiently open cryptic pockets and identified 12 new STEP inhibition scaffolds through structure-based virtual screening. Site-directed mutagenesis verified the binding of ST3 with the predicted cryptic pockets. Moreover, the most potent and selective inhibitors could modulate the phosphorylation of both ERK1/2 and Pyk2 in PC12 cells.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Furanos/farmacología , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Quinolinas/farmacología , Animales , Sitios de Unión , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Furanos/química , Furanos/metabolismo , Humanos , Simulación de Dinámica Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Mutación , Células PC12 , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Ratas , Relación Estructura-Actividad
6.
J Cell Biochem ; 121(5-6): 3298-3312, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31898344

RESUMEN

Protein tyrosine phosphatase non-receptor type 21 (PTPN21) is a member of the non-receptor tyrosine phosphatase family. We have found that PTPN21 is mutated in relapsed Philadelphia chromosome-negative acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem cell transplantation. PTPN21 consists of three types of isoforms according to the length of the protein encoded. However, the roles of different isoforms in leukemic cells have not been elucidated. In the study, PTPN21 isoform constitution in five ALL cell lines were identified by transcriptome polymerase chain reaction combined with Sanger sequencing, and the relationship between PTPN21 isoforms and sensitivity to natural killer (NK) cells mediated killing in ALL cell lines were further assessed by knock-out of different isoforms of PTPN21 using CRISPR-Cas9 technique. Subsequently, we explored the functional mechanisms through RNA sequencing and confirmatory testing. The results showed that there was no significant change when all PTPN21 isoforms were knocked out in ALL cells, but the sensitivity of NALM6 cells with PTPN21-CDSlong knock-out (NALM6-PTPN21lk ) to NK-mediated killing was significantly increased. Whole transcriptome sequencing and further validation testing showed that human leukocyte antigen class I (HLA-I) molecules were significantly decreased, accompanied by a significantly downregulated expression of antigen presenting-related chaperones in NALM6-PTPN21lk cells. Our results uncovered a previously unknown mechanism that PTPN21-CDSlong and CDSshort isoforms may play opposite roles in NK-mediated killing in ALL cells, and showed that the endogenous PTPN21-CDSlong isoform inhibited ALL cells to NK cell-mediated lysis by regulating the KIR-HLA-I axis.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Sistemas CRISPR-Cas , Muerte Celular , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Edición Génica , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Chaperonas Moleculares/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Isoformas de Proteínas , RNA-Seq
7.
J Virol ; 94(4)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31748394

RESUMEN

Capsid envelopment during assembly of the neurotropic herpesviruses herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV) in the infected cell cytoplasm is thought to involve the late-acting cellular ESCRT (endosomal sorting complex required for transport) components ESCRT-III and VPS4 (vacuolar protein sorting 4). However, HSV-1, unlike members of many other families of enveloped viruses, does not appear to require the ESCRT-I subunit TSG101 or the Bro1 domain-containing protein ALIX (Alg-2-interacting protein X) to recruit and activate ESCRT-III. Alternative cellular factors that are known to be capable of regulating ESCRT-III function include the ESCRT-II complex and other members of the Bro1 family. We therefore used small interfering RNA (siRNA) to knock down the essential ESCRT-II subunit EAP20/VPS25 (ELL-associated protein 20/vacuolar protein sorting 25) and the Bro1 proteins HD-PTP (His domain-containing protein tyrosine phosphatase) and BROX (Bro1 domain and CAAX motif containing). We demonstrated reductions in levels of the targeted proteins by Western blotting and used quantitative microscopic assays to confirm loss of ESCRT-II and HD-PTP function. We found that in single-step replication experiments, the final yields of HSV-1 were unchanged following loss of EAP20, HD-PTP, or BROX.IMPORTANCE HSV-1 is a pathogen of the human nervous system that uses its own virus-encoded proteins and the normal cellular ESCRT machinery to drive the construction of its envelope. How HSV-1 structural proteins interact with ESCRT components and which subsets of cellular ESCRT proteins are utilized by the virus remain largely unknown. Here, we demonstrate that an essential component of the ESCRT-II complex and two ESCRT-associated Bro1 proteins are dispensable for HSV-1 replication.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cápside/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/metabolismo , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Unión Proteica/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/genética , Interferencia de ARN , Proteínas de Transporte Vesicular/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
8.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118613, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31758957

RESUMEN

Lafora progressive myoclonus epilepsy is a fatal rare neurodegenerative disorder characterized by the accumulation of insoluble abnormal glycogen deposits in the brain and peripheral tissues. Mutations in at least two genes are responsible for the disease: EPM2A, encoding the glucan phosphatase laforin, and EPM2B, encoding the RING-type E3-ubiquitin ligase malin. Both laforin and malin form a functional complex in which laforin recruits the substrates to be ubiquitinated by malin. We and others have described that, in cellular and animal models of this disease, there is an autophagy impairment which leads to the accumulation of dysfunctional mitochondria. In addition, we established that the autophagic defect occurred at the initial steps of autophagosome formation. In this work, we present evidence that in cellular models of the disease there is a decrease in the amount of phosphatidylinositol-3P. This is probably due to defective regulation of the autophagic PI3KC3 complex, in the absence of a functional laforin/malin complex. In fact, we demonstrate that the laforin/malin complex interacts physically and co-localizes intracellularly with core components of the PI3KC3 complex (Beclin1, Vps34 and Vps15), and that this interaction is specific and results in the polyubiquitination of these proteins. In addition, the laforin/malin complex is also able to polyubiquitinate ATG14L and UVRAG. Finally, we show that overexpression of the laforin/malin complex increases PI3KC3 activity. All these results suggest a new role of the laforin/malin complex in the activation of autophagy via regulation of the PI3KC3 complex and explain the defect in autophagy described in Lafora disease.


Asunto(s)
Enfermedad de Lafora/patología , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Autofagia , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/química , Beclina-1/metabolismo , Células Cultivadas , Humanos , Enfermedad de Lafora/metabolismo , Microscopía Fluorescente , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/genética , Factores de Transcripción/química , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
9.
PLoS Biol ; 17(7): e3000367, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31323018

RESUMEN

Human papillomaviruses (HPVs) are causative agents of various diseases associated with cellular hyperproliferation, including cervical cancer, one of the most prevalent tumors in women. E7 is one of the two HPV-encoded oncoproteins and directs recruitment and subsequent degradation of tumor-suppressive proteins such as retinoblastoma protein (pRb) via its LxCxE motif. E7 also triggers tumorigenesis in a pRb-independent pathway through its C-terminal domain, which has yet been largely undetermined, with a lack of structural information in a complex form with a host protein. Herein, we present the crystal structure of the E7 C-terminal domain of HPV18 belonging to the high-risk HPV genotypes bound to the catalytic domain of human nonreceptor-type protein tyrosine phosphatase 14 (PTPN14). They interact directly and potently with each other, with a dissociation constant of 18.2 nM. Ensuing structural analysis revealed the molecular basis of the PTPN14-binding specificity of E7 over other protein tyrosine phosphatases and also led to the identification of PTPN21 as a direct interacting partner of E7. Disruption of HPV18 E7 binding to PTPN14 by structure-based mutagenesis impaired E7's ability to promote keratinocyte proliferation and migration. Likewise, E7 binding-defective PTPN14 was resistant for degradation via proteasome, and it was much more effective than wild-type PTPN14 in attenuating the activity of downstream effectors of Hippo signaling and negatively regulating cell proliferation, migration, and invasion when examined in HPV18-positive HeLa cells. These results therefore demonstrated the significance and therapeutic potential of the intermolecular interaction between HPV E7 and host PTPN14 in HPV-mediated cell transformation and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Femenino , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Unión Proteica , Dominios Proteicos , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Homología de Secuencia de Aminoácido , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
10.
Mol Genet Genomic Med ; 7(5): e621, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30884204

RESUMEN

BACKGROUND: X-linked myotubular myopathy (XLMTM) is a form of the severest congenital muscle diseases characterized by marked muscle weakness, hypotonia, and feeding and breathing difficulties in male infants. It is caused by mutations in the myotubularin gene (MTM1). METHODS: Evaluation of clinical history and examination of muscle pathology of three patients and comprehensive genome analysis on our original targeted gene panel system for muscular diseases. RESULTS: We report three patients, each of whom presents distinct muscle pathological features. The three patients have novel hemizygous MTM1 variants, including c.527A>G (p.Gln176Arg), c.595C>G (p.Pro199Ala), or c.688T>C (p.Trp230Arg). CONCLUSIONS: All variants were assessed as "Class 4 (likely pathogenic)" on the basis of the guideline of American College of Medical Genetics and Genomics. These distinct pathological features among the patients with variants in the second cluster of PTP domain in MTM1 provides an insight into microheterogeneities in disease phenotypes in XLMTM.


Asunto(s)
Mutación Missense , Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Adolescente , Niño , Hemicigoto , Humanos , Lactante , Recién Nacido , Masculino , Miopatías Estructurales Congénitas/patología , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
11.
Biochem Cell Biol ; 97(1): 68-72, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29879361

RESUMEN

Cell surface receptors trigger the activation of signaling pathways to regulate key cellular processes, including cell survival and proliferation. Internalization, sorting, and trafficking of activated receptors, therefore, play a major role in the regulation and attenuation of cell signaling. Efficient sorting of endocytosed receptors is performed by the ESCRT machinery, which targets receptors for degradation by the sequential establishment of protein complexes. These events are tightly regulated and malfunction of ESCRT components can lead to abnormal trafficking and sustained signaling and promote tumor formation or progression. In this review, we analyze the modular domain organization of the alternative ESCRT protein HD-PTP and its role in receptor trafficking and tumorigenesis.


Asunto(s)
Endocitosis/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Neoplasias/fisiopatología , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Humanos , Transporte de Proteínas , Relación Estructura-Actividad
12.
J Med Chem ; 62(1): 306-316, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30207464

RESUMEN

Protein tyrosine phosphatase non-receptor type 5 (PTPN5, STEP) is a brain specific phosphatase that regulates synaptic function and plasticity by modulation of N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking. Dysregulation of STEP has been linked to neurodegenerative and neuropsychiatric diseases, highlighting this enzyme as an attractive therapeutic target for drug discovery. Selective targeting of STEP with small molecules has been hampered by high conservation of the active site among protein tyrosine phosphatases. We report the discovery of the first small molecule allosteric activator for STEP that binds to the phosphatase domain. Allosteric binding is confirmed by both X-ray and 15N NMR experiments, and specificity has been demonstrated by an enzymatic test cascade. Molecular dynamics simulations indicate stimulation of enzymatic activity by a long-range allosteric mechanism. To allow the scientific community to make use of this tool, we offer to provide the compound in the course of an open innovation initiative.


Asunto(s)
Proteínas Tirosina Fosfatasas no Receptoras/química , Bibliotecas de Moléculas Pequeñas/química , Regulación Alostérica , Sitio Alostérico , Animales , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Ratones , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo
13.
Anal Biochem ; 563: 51-55, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30291838

RESUMEN

Glucan phosphatases are a unique subset of the phosphatase family that bind to and dephosphorylate carbohydrate substrates. Family members are found in diverse organisms ranging from single-cell red algae to humans. The nature of their functional oligomerization has been a source of considerable debate. We demonstrate that the human laforin protein behaves aberrantly when subjected to Size Exclusion Chromotography (SEC) analysis due to interaction with the carbohydrate-based matrix. This interaction complicates the analysis of laforin human disease mutations. Herein, we show that SEC with Multi-Angle static Light Scattering (SEC-MALS) provides a method to robustly define the oligomerization state of laforin and laforin variants. We further analyzed glucan phosphatases from photosynthetic organisms to define if this interaction was characteristic of all glucan phosphatases. Starch EXcess-four (SEX4) from green plants was found to lack significant interaction with the matrix and instead exists as a monomer. Conversely, Cm-laforin, from red algae, exists as a monomer in solution while still exhibiting significant interaction with the matrix. These data demonstrate a range of oligomerization behaviors of members of the glucan phosphatase family, and establish SEC-MALS as a robust methodology to quantify and compare oligomerization states between different proteins and protein variants in this family.


Asunto(s)
Carbohidratos/química , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Cromatografía en Gel , Glucanos/metabolismo , Humanos , Unión Proteica , Multimerización de Proteína
14.
FEBS Lett ; 592(4): 586-598, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29389008

RESUMEN

Laforin catalyses glycogen dephosphorylation. Mutations in its gene result in Lafora disease, a fatal progressive myoclonus epilepsy, the hallmark being water-insoluble, hyperphosphorylated carbohydrate inclusions called Lafora bodies. Human laforin consists of an N-terminal carbohydrate-binding module (CBM) from family CBM20 and a C-terminal dual-specificity phosphatase domain. Laforin is conserved in all vertebrates, some basal metazoans and a small group of protozoans. The present in silico study defines the evolutionary relationships among the CBM20s of laforin with an emphasis on newly identified laforin orthologues. The study reveals putative laforin orthologues in Trichinella, a parasitic nematode, and identifies two sequence inserts in the CBM20 of laforin from parasitic coccidia. Finally, we identify that the putative laforin orthologues from some protozoa and algae possess more than one CBM20.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Evolución Molecular , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Simulación por Computador , Humanos , Dominios Proteicos
15.
Nat Cell Biol ; 20(2): 198-210, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29358706

RESUMEN

The ubiquitin proteasome system and autophagy are major protein turnover mechanisms in muscle cells, which ensure stemness and muscle fibre maintenance. Muscle cells contain a high proportion of cytoskeletal proteins, which are prone to misfolding and aggregation; pathological processes that are observed in several neuromuscular diseases called proteinopathies. Despite advances in deciphering the mechanisms underlying misfolding and aggregation, little is known about how muscle cells manage cytoskeletal degradation. Here, we describe a process by which muscle cells degrade the misfolded intermediate filament proteins desmin and vimentin by the proteasome. This relies on the MTM1-UBQLN2 complex to recognize and guide these misfolded proteins to the proteasome and occurs prior to aggregate formation. Thus, our data highlight a safeguarding function of the MTM1-UBQLN2 complex that ensures cytoskeletal integrity to avoid proteotoxic aggregate formation.


Asunto(s)
Autofagia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Filamentos Intermediarios/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Ubiquitinas/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas Relacionadas con la Autofagia , Proteínas de Ciclo Celular/química , Citoesqueleto/genética , Desmina/genética , Humanos , Proteínas de Filamentos Intermediarios/química , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Agregado de Proteínas/genética , Pliegue de Proteína , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteolisis , Ubiquitina/genética , Ubiquitinas/química , Vimentina/genética
16.
Mol Neurobiol ; 55(4): 3546-3550, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28509084

RESUMEN

Charcot-Marie-Tooth (CMT) disease refers to a heterogeneous group of axonal and demyelinating polyneuropathies, characterized by chronic motor and sensory dysfunction. CMT is the most common genetic cause of neuropathy. The present study aimed to identify the gene mutation responsible for CMT in Ashkenazi Jew (AJ) patient. Genomic DNA was extracted from whole blood leukocytes of affected family and normal subject. Whole-exome sequencing was performed using the Illumina HiSeq2500. The DNA region containing the identified mutation was amplified by PCR and sequenced using dye-terminator chemistry and the forward primer. Physical examination of the patient revealed weakness and atrophy of the lower extremity muscles and Pes cavus foot deformity. Whole-exome sequencing indicated that the patient is homozygous for a novel frameshift mutation (c.1877_1878insAGAG, p.Arg630fs) in the myotubularin-related protein-2 gene (MTMR2), which resulted in an erroneous C-terminal sequence and extension by 15 amino acids. Patients' parents are healthy, and DNA sequencing analysis indicated that both are heterozygotes to the described mutation. The clinical feature of the patient may indicate a complete co-segregation of the p.Arg630fs mutation in MTMR2 gene with the CMT type 4B1 phenotype. Further studies are needed in order to estimate the prevalence of this mutation among AJ.


Asunto(s)
Secuenciación del Exoma , Mutación del Sistema de Lectura/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Enfermedad de Charcot-Marie-Tooth , Niño , Femenino , Homocigoto , Humanos , Dominios Proteicos , Proteínas Tirosina Fosfatasas no Receptoras/química
17.
J Med Chem ; 60(22): 9299-9319, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29116812

RESUMEN

Excessive activity of striatal-enriched protein tyrosine phosphatase (STEP) in the brain has been detected in numerous neuropsychiatric disorders including Alzheimer's disease. Notably, knockdown of STEP in an Alzheimer mouse model effected an increase in the phosphorylation levels of downstream STEP substrates and a significant reversal in the observed cognitive and memory deficits. These data point to the promising potential of STEP as a target for drug discovery in Alzheimer's treatment. We previously reported a substrate-based approach to the development of low molecular weight STEP inhibitors with Ki values as low as 7.8 µM. Herein, we disclose the first X-ray crystal structures of inhibitors bound to STEP and the surprising finding that they occupy noncoincident binding sites. Moreover, we utilize this structural information to optimize the inhibitor structure to achieve a Ki of 110 nM, with 15-60-fold selectivity across a series of phosphatases.


Asunto(s)
Organofosfonatos/química , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Sulfonamidas/química , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Dominio Catalítico , Cristalografía por Rayos X , Descubrimiento de Drogas , Estabilidad de Medicamentos , Fosfatasas de Especificidad Dual/antagonistas & inhibidores , Microsomas Hepáticos/metabolismo , Organofosfonatos/síntesis química , Organofosfonatos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/química , Ratas , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo
18.
Hum Genet ; 136(11-12): 1455-1461, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29090338

RESUMEN

Developmental and epileptic encephalopathies (DEE) are a heterogeneous group of neurodevelopmental disorders with poor prognosis. Recent discoveries have greatly expanded the repertoire of genes that are mutated in epileptic encephalopathies and DEE, often in a de novo fashion, but in many patients, the disease remains molecularly uncharacterized. Here, we describe a new form of DEE in patients with likely deleterious biallelic variants in PTPN23. The phenotype is characterized by early onset drug-resistant epilepsy, severe and global developmental delay, microcephaly, and sometimes premature death. PTPN23 encodes a tyrosine phosphatase with strong brain expression, and its knockout in mouse is embryonically lethal. Structural modeling supports a deleterious effect of the identified alleles. Our data suggest that PTPN23 mutations cause a rare severe form of autosomal-recessive DEE in humans, a finding that requires confirmation.


Asunto(s)
Discapacidades del Desarrollo/genética , Mutación , Proteínas Tirosina Fosfatasas no Receptoras/genética , Espasmos Infantiles/genética , Adulto , Discapacidades del Desarrollo/patología , Femenino , Humanos , Recién Nacido , Masculino , Fenotipo , Conformación Proteica , Proteínas Tirosina Fosfatasas no Receptoras/química , Espasmos Infantiles/patología
19.
Sci Rep ; 7(1): 9151, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28831121

RESUMEN

HD-PTP is a tumour suppressor phosphatase that controls endocytosis, down-regulation of mitogenic receptors and cell migration. Central to its role is the specific recruitment of critical endosomal sorting complexes required for transport (ESCRTs). However, the molecular mechanisms that enable HD-PTP to regulate ESCRT function are unknown. We have characterised the molecular architecture of the entire ESCRT binding region of HD-PTP using small angle X-ray scattering and hydrodynamic analyses. We show that HD-PTP adopts an open and extended conformation, optimal for concomitant interactions with multiple ESCRTs, which contrasts with the compact conformation of the related ESCRT regulator Alix. We demonstrate that the HD-PTP open conformation is functionally competent for binding cellular protein partners. Our analyses rationalise the functional cooperation of HD-PTP with ESCRT-0, ESCRT-I and ESCRT-III and support a model for regulation of ESCRT function by displacement of ESCRT subunits, which is crucial in determining the fate of ubiquitinated cargo.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Hidrodinámica , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
20.
Structure ; 25(7): 1011-1024.e4, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28602823

RESUMEN

SARA and endofin are endosomal adaptor proteins that drive Smad phosphorylation by ligand-activated transforming growth factor ß/bone morphogenetic protein (TGFß/BMP) receptors. We show in this study that SARA and endofin also recruit the tumor supressor HD-PTP, a master regulator of endosomal sorting and ESCRT-dependent receptor downregulation. High-affinity interactions occur between the SARA/endofin N termini, and the conserved hydrophobic region in the HD-PTP Bro1 domain that binds CHMP4/ESCRT-III. CHMP4 engagement is a universal feature of Bro1 proteins, but SARA/endofin binding is specific to HD-PTP. Crystallographic structures of HD-PTPBro1 in complex with SARA, endofin, and three CHMP4 isoforms revealed that all ligands bind similarly to the conserved site but, critically, only SARA/endofin interact at a neighboring pocket unique to HD-PTP. The structures, together with mutagenesis and binding analysis, explain the high affinity and specific binding of SARA/endofin, and why they compete so effectively with CHMP4. Our data invoke models for how endocytic regulation of TGFß/BMP signaling is controlled.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Simulación del Acoplamiento Molecular , Proteínas Tirosina Fosfatasas no Receptoras/química , Serina Endopeptidasas/química , Sitios de Unión , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Serina Endopeptidasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...