Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Genet ; 1032024.
Artículo en Inglés | MEDLINE | ID: mdl-38562040

RESUMEN

Intellectual developmental disorder, X-linked 104 (XLID104), caused by the FRMPD4 gene variant, is a rare X-linked genetic disease that primarily manifests as intellectual disability (ID) and language delay, and may be accompanied by behavioural abnormalities. Currently, only 11 patients from four families have been reported to carry FRMPD4 gene variants. Here, we report a rare case of a Chinese patient with XLID104 who was presented with severe ID and language impairment. Genetic testing results showed that the patient had a novel hemizygous variant on FRMPD4 inherited from the heterozygous variant NM_001368397: c.1772A>C (p.Glu591Ala) carried by his mother. To our knowledge, this variant has not been reported previously. Western blot results for the recombinant plasmid constructed in vitro indicated that the expression of the mutant protein may be reduced. Using molecular dynamics simulations, we predicted that the mutant protein may affect the interaction of the FRMPD4 protein with DLG4. In this study, we expand the spectrum of FRMPD4 variants and suggest that the clinical awareness of the genetic diagnosis of nonsyndromic ID should be strengthened.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X , Discapacidad Intelectual , Niño , Humanos , Discapacidad Intelectual/genética , Dominios FERM , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Proteínas Mutantes/genética
2.
Sci Adv ; 10(9): eadi7404, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38416831

RESUMEN

PTPN21 belongs to the four-point-one, ezrin, radixin, moesin (FERM) domain-containing protein tyrosine phosphatases (PTP) and plays important roles in cytoskeleton-associated cellular processes like cell adhesion, motility, and cargo transport. Because of the presence of a WPE loop instead of a WPD loop in the phosphatase domain, it is often considered to lack phosphatase activity. However, many of PTPN21's biological functions require its catalytic activity. To reconcile these findings, we have determined the structures of individual PTPN21 FERM, PTP domains, and a complex between FERM-PTP. Combined with biochemical analysis, we have found that PTPN21 PTP is weakly active and is autoinhibited by association with its FERM domain. Disruption of FERM-PTP interaction results in enhanced ERK activation. The oncogenic HPV18 E7 protein binds to PTP at the same location as PTPN21 FERM, indicating that it may act by displacing the FERM domain from PTP. Our results provide mechanistic insight into PTPN21 and benefit functional studies of PTPN21-mediated processes.


Asunto(s)
Dominios FERM , Proteínas Tirosina Fosfatasas , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas/metabolismo , Unión Proteica , Citoesqueleto/metabolismo
3.
Life Sci Alliance ; 7(4)2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38296350

RESUMEN

The FERM domain is a conserved and widespread protein module that appeared in the common ancestor of amoebae, fungi, and animals, and is therefore now found in a wide variety of species. The primary function of the FERM domain is localizing to the plasma membrane through binding lipids and proteins of the membrane; thus, for a long time, FERM domain-containing proteins (FDCPs) were considered exclusively cytoskeletal. Although their role in the cytoplasm has been extensively studied, the recent discovery of the presence and importance of cytoskeletal proteins in the nucleus suggests that FDCPs might also play an important role in nuclear function. In this review, we collected data on their nuclear localization, transport, and possible functions, which are still scattered throughout the literature, with special regard to the role of the FERM domain in these processes. With this, we would like to draw attention to the exciting, new dimension of the role of FDCPs, their nuclear activity, which could be an interesting novel direction for future research.


Asunto(s)
Proteínas del Citoesqueleto , Dominios FERM , Animales , Estructura Terciaria de Proteína , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo
4.
J Thromb Haemost ; 22(2): 558-564, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37866516

RESUMEN

BACKGROUND: Leukocyte adhesion deficiency III (LAD-III) is a rare autosomal recessive syndrome characterized by functional deficiencies of platelets and leukocytes that occurs due to mutations in the FERMT3 gene encoding kindlin-3. Kindlin-3 is a FERM domain-containing adaptor protein that is essential in integrin activation. We have previously demonstrated that the FERM domain of kindlin-3 is structurally compact and plays an important role in supporting integrin activation in a mouse model. The impact of destabilizing the compact FERM domain in kindlin-3 on the development of LAD-III in humans remains uncertain. OBJECTIVES: To use primary cells from a patient with LAD-III to validate the role of the compact FERM domain in kindlin-3 function in platelets and leukocytes. METHODS: The patient is a 4-year-old girl who since infancy has displayed clinical features of LAD-III. Patient platelets and leukocytes were functionally analyzed, and structural analysis of the kindlin-3 variant was conducted. RESULTS: We identified a novel homozygous missense mutation in the FERMT3 (c.412G>A, p.E138K) FERM domain. Substantially reduced levels of kindlin-3 were detected in the proband's platelets and leukocytes. Functional evaluation verified that integrin αIIbß3-mediated platelet activation, spreading, and aggregation and ß2-integrin-mediated neutrophil adhesion and spreading were significantly compromised. Structural analysis revealed that this newly identified E138K substitution in kindlin-3 destabilizes the compacted FERM domain, resulting in poor expression of kindlin-3 in blood cells and subsequent LAD-III. CONCLUSION: We have identified a novel missense mutation and verified the functional significance of the compact kindlin-3 FERM domain in supporting integrin functions in platelets and leukocytes.


Asunto(s)
Dominios FERM , Síndrome de Deficiencia de Adhesión del Leucocito , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Animales , Preescolar , Femenino , Humanos , Ratones , Adhesión Celular/genética , Proteínas del Citoesqueleto/metabolismo , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Leucocitos/metabolismo , Mutación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo
5.
J Biol Chem ; 299(12): 105382, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37866628

RESUMEN

Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.


Asunto(s)
Enfermedad de Alzheimer , Unión Proteica , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Dominios FERM , Receptores de Hialuranos/metabolismo , Unión Proteica/efectos de los fármacos , Proteómica
6.
Life Sci Alliance ; 6(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37280085

RESUMEN

NF2 (moesin-ezrin-radixin-like [MERLIN] tumor suppressor) is frequently inactivated in cancer, where its NF2 tumor suppressor functionality is tightly coupled to protein conformation. How NF2 conformation is regulated and how NF2 conformation influences tumor suppressor activity is a largely open question. Here, we systematically characterized three NF2 conformation-dependent protein interactions utilizing deep mutational scanning interaction perturbation analyses. We identified two regions in NF2 with clustered mutations which affected conformation-dependent protein interactions. NF2 variants in the F2-F3 subdomain and the α3H helix region substantially modulated NF2 conformation and homomerization. Mutations in the F2-F3 subdomain altered proliferation in three cell lines and matched patterns of disease mutations in NF2 related-schwannomatosis. This study highlights the power of systematic mutational interaction perturbation analysis to identify missense variants impacting NF2 conformation and provides insight into NF2 tumor suppressor function.


Asunto(s)
Neoplasias , Neurofibromina 2 , Humanos , Neurofibromina 2/genética , Neurofibromina 2/química , Neurofibromina 2/metabolismo , Dominios FERM , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Conformación Proteica
7.
Biochemistry ; 62(11): 1594-1607, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37224425

RESUMEN

The ERM (ezrin, radixin, and moesin) family of proteins and the related protein merlin participate in scaffolding and signaling events at the cell cortex. The proteins share an N-terminal FERM [band four-point-one (4.1) ERM] domain composed of three subdomains (F1, F2, and F3) with binding sites for short linear peptide motifs. By screening the FERM domains of the ERMs and merlin against a phage library that displays peptides representing the intrinsically disordered regions of the human proteome, we identified a large number of novel ligands. We determined the affinities for the ERM and merlin FERM domains interacting with 18 peptides and validated interactions with full-length proteins through pull-down experiments. The majority of the peptides contained an apparent Yx[FILV] motif; others show alternative motifs. We defined distinct binding sites for two types of similar but distinct binding motifs (YxV and FYDF) using a combination of Rosetta FlexPepDock computational peptide docking protocols and mutational analysis. We provide a detailed molecular understanding of how the two types of peptides with distinct motifs bind to different sites on the moesin FERM phosphotyrosine binding-like subdomain and uncover interdependencies between the different types of ligands. The study expands the motif-based interactomes of the ERMs and merlin and suggests that the FERM domain acts as a switchable interaction hub.


Asunto(s)
Dominios FERM , Neurofibromina 2 , Humanos , Neurofibromina 2/genética , Neurofibromina 2/química , Neurofibromina 2/metabolismo , Ligandos , Estructura Terciaria de Proteína , Péptidos
8.
Exp Anim ; 72(3): 324-335, 2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-36740252

RESUMEN

Intracerebral hemorrhage (ICH) is an incurable neurological disease. Microglia activation and its related inflammation contribute to ICH-associated brain damage. FERM domain containing kindlin 1 (FERMT1) is an integrin-binding protein that participates in microglia-associated inflammation, but its role in ICH is unclear. An ICH model was constructed by injecting 50 µl of autologous blood into the bregma of rats. FERMT1 siRNA was injected into the right ventricle of the rat for knockdown of FERMT1. A significant striatal hematoma was observed in ICH rats. FERMT1 knockdown reduced the water content of brain tissue, alleviated brain hematoma and improved behavioral function in ICH rats. FERMT1 knockdown reduced microglia activity, inhibited NLR family pyrin domain containing 3 (NLRP3) inflammasome activity and decreased the expression of inflammatory factors including IL-1ß and IL-18 in the peri-hematoma tissues. BV2 microglial cells were transfected with FERMT1 siRNA and incubated with 60 µM Hemin for 24 h. Activation of NLRP3 inflammasome induced by hemin were reduced in microglia when FERMT1 was knocked down, leading to decreased production of inflammatory factors IL-1ß and IL-18. In addition, knockdown of FERMT1 prevented the activation of nuclear factor kappa B (NF-κB) signaling pathway in vivo and in vitro. Our findings suggested that down-regulation of FERMT1 attenuated microglial inflammation and brain damage induced by ICH via NLRP3/NF-κB pathway. FERMT1 is a key regulator of inflammatory damage in rats after ICH.


Asunto(s)
Lesiones Encefálicas , FN-kappa B , Animales , Ratas , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral , Dominios FERM , Hemina , Inflamasomas/metabolismo , Inflamación , Interleucina-18 , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dominio Pirina , ARN Interferente Pequeño
9.
Biophys J ; 122(7): 1325-1333, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36814382

RESUMEN

The four-point-one ezrin-radixin-moesin homology (FERM) protein domain is a multifunctional protein-lipid binding site, constituting an integral part of numerous membrane-associated proteins. Its interaction with the lipid phosphatidylinositol-4,5-bisphosphate (PIP2), located at the inner leaflet of eukaryotic plasma membranes, is important for localization, anchorage, and activation of FERM-containing proteins. FERM-PIP2 complexes structurally determined so far exclusively feature a 1:1 binding stoichiometry of protein and lipid, with a few basic FERM residues neutralizing the -4 charge of the bound PIP2. Whether this picture from static crystal structures also applies to the dynamic interaction of FERM domains on PIP2 membranes is unknown. We here quantified the stoichiometry of FERM-PIP2 binding in a lipid bilayer using atomistic molecular dynamics simulations and experiments on solid supported membranes for the FERM domains of focal adhesion kinase and ezrin. In contrast to the structural data, we find much higher average stoichiometries of FERM-PIP2 binding, amounting to 1:3 or 1:4 ratios, respectively. In simulations, the full set of basic residues at the membrane interface, 7 and 15 residues for focal adhesion kinase and ezrin, respectively, engages in PIP2 interactions. In addition, Na ions enter the FERM-membrane binding interface, compensating negative PIP2 charges in case of high charge surpluses from bound PIP2. We propose the multivalent binding of FERM domains to PIP2 in lipid bilayers to significantly enhance the stability of FERM-membrane binding and to render the FERM-membrane linkage highly adjustable.


Asunto(s)
Dominios FERM , Membrana Dobles de Lípidos , Sitios de Unión , Membrana Celular/metabolismo , Unión Proteica , Membrana Dobles de Lípidos/química , Proteína-Tirosina Quinasas de Adhesión Focal/química , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo
10.
Biophys J ; 122(7): 1277-1286, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36814383

RESUMEN

Focal adhesions (FAs) mediate the interaction of the cytoskeleton with the extracellular matrix in a highly dynamic fashion. Talin is a central regulator, adaptor protein, and mechano-sensor of FA complexes. For recruitment and firm attachment at FAs, talin's N-terminal FERM domain binds to phosphatidylinositol 4,5-bisphosphate (PIP2)-enriched membranes. A newly published autoinhibitory structure of talin-1, where the known PIP2 interaction sites are covered up, lead us to hypothesize that a hitherto less examined loop insertion of the FERM domain acts as an additional and initial site of contact. We evaluated direct interactions of talin-1 with a PIP2 membrane by means of atomistic molecular dynamics simulations. We show that this unstructured, 33-residue-long loop strongly interacts with PIP2 and can facilitate further membrane contacts, including the canonical PIP2 interactions, by serving as a flexible membrane anchor. Under force as present at FAs, the extensible FERM loop ensures talin maintains membrane contacts when pulled away from the membrane by up to 7 nm. We identify key basic residues of the anchor mediating the highly dynamic talin-membrane interaction. Our results put forward an intrinsically disordered loop as a key and highly adaptable PIP2 recognition site of talin and potentially other PIP2-binding mechano-proteins.


Asunto(s)
Dominios FERM , Talina , Talina/metabolismo , Adhesiones Focales/metabolismo , Citoesqueleto/metabolismo , Simulación de Dinámica Molecular , Proteínas Portadoras/metabolismo , Unión Proteica , Sitios de Unión
11.
J Invest Dermatol ; 142(9): 2415-2423.e1, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35189150

RESUMEN

Kindler epidermolysis bullosa is a genodermatosis that manifests with cutaneous and mucosal fragility and with photosensitivity. No cure is available to date. Kindlin-1, a deficient protein, binds to ß-integrin and is required for its activation. Using a previously established experimental workflow, we addressed the consequences of three naturally occurring pathogenic variants, leading either to single amino acid substitutions p.Y293D and p.W559R or to a single amino acid deletion p.I623del in kindlin-1. We show that p.Y293D disrupts kindlin-1 localization to focal adhesions and cell spreading. Although treatment with a chemical chaperone increases the amount of mutant protein, spreading does not improve, and cellular stress increases, whereas the variants p.W559R and p.I623del do not interfere with kindlin-1 localization to focal adhesions and support cell adhesion and survival. These mutants are also responsive to the treatment with a chemical chaperone, and the increased mutant proteins improve cell spreading. These findings suggest that low levels of mutant kindlins p.W559R and p.I623del are able to rescue some important cellular functions. Patients carrying these mutations could benefit from treatment with promotors of proteostasis. Our results show that each pathogenic variant must be individually tested on genetic, molecular, and cellular levels to tailor personalized treatments for patients.


Asunto(s)
Dominios FERM , Proteínas de la Membrana , Proteínas de Neoplasias , Medicina de Precisión , Sustitución de Aminoácidos , Humanos , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética
12.
Biofactors ; 48(2): 428-441, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34669997

RESUMEN

The emerging role of FERM domain-containing protein 6 (FRMD6) in cancer progression has been revealed in several malignancies. However, its relevance on thyroid cancer is not well understood. This work evaluated the possible role and mechanism of FRMD6 in thyroid cancer. We demonstrated that FRMD6 expression was downregulated in thyroid cancer by analyzing the Cancer Genome Atlas data. Remarkable reductions in FRMD6 expression were also confirmed in the clinical specimens and cell lines of thyroid cancer. The upregulation of FRMD6 restrained the proliferation, epithelial-mesenchymal transition, and invasion of thyroid cancer. Moreover, FRMD6 overexpression significantly increased the apoptosis and cell cycle arrest. Further molecular research demonstrated that the overexpression of FRMD6 increased the phosphorylation levels of mammalian STE20-like protein kinase 1, large tumor suppressor 1, and Yes-associated protein 1 (YAP1) and prohibited the activation of YAP1. The re-expression of constitutively active YAP1 strikingly reversed FRMD6-induced tumor-inhibiting effects. Thyroid cancer cells overexpressing FRMD6 had a weakened ability to form xenograft tumors in vivo in nude mice. Overall, the overexpression of FRMD6 produces remarkable tumor-inhibiting effects in thyroid cancer by inhibiting oncogenic YAP1.


Asunto(s)
Dominios FERM , Neoplasias de la Tiroides , Animales , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Mamíferos , Ratones , Ratones Desnudos , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Proteínas Señalizadoras YAP
13.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34462354

RESUMEN

The molecular events that determine the recycling versus degradation fates of internalized membrane proteins remain poorly understood. Two of the three members of the SNX-FERM family, SNX17 and SNX31, utilize their FERM domain to mediate endocytic trafficking of cargo proteins harboring the NPxY/NxxY motif. In contrast, SNX27 does not recycle NPxY/NxxY-containing cargo but instead recycles cargo containing PDZ-binding motifs via its PDZ domain. The underlying mechanism governing this divergence in FERM domain binding is poorly understood. Here, we report that the FERM domain of SNX27 is functionally distinct from SNX17 and interacts with a novel DLF motif localized within the N terminus of SNX1/2 instead of the NPxY/NxxY motif in cargo proteins. The SNX27-FERM-SNX1 complex structure reveals that the DLF motif of SNX1 binds to a hydrophobic cave surrounded by positively charged residues on the surface of SNX27. The interaction between SNX27 and SNX1/2 is critical for efficient SNX27 recruitment to endosomes and endocytic recycling of multiple cargoes. Finally, we show that the interaction between SNX27 and SNX1/2 is critical for brain development in zebrafish. Altogether, our study solves a long-standing puzzle in the field and suggests that SNX27 and SNX17 mediate endocytic recycling through fundamentally distinct mechanisms.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Dominios FERM , Nexinas de Clasificación/metabolismo , Animales , Encéfalo/metabolismo , Endocitosis , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Neuronas/citología , Unión Proteica , Transporte de Proteínas , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Nexinas de Clasificación/química , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
14.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32992803

RESUMEN

The PSGL-1-actin cytoskeleton linker proteins ezrin/radixin/moesin (ERM), an adaptor between P-selectin glycoprotein ligand-1 (PSGL-1) and spleen tyrosine kinase (Syk), is a key player in PSGL-1 signal, which mediates the adhesion and recruitment of leukocytes to the activated endothelial cells in flow. Binding of PSGL-1 to ERM initials intracellular signaling through inducing phosphorylation of Syk, but effects of tensile force on unligation and phosphorylation site exposure of ERM bound with PSGL-1 remains unclear. To answer this question, we performed a series of so-called "ramp-clamp" steered molecular dynamics (SMD) simulations on the radixin protein FERM domain of ERM bound with intracellular juxtamembrane PSGL-1 peptide. The results showed that, the rupture force of complex pulled with constant velocity was over 250 pN, which prevented the complex from breaking in front of pull-induced exposure of phosphorylation site on immunoreceptor tyrosine activation motif (ITAM)-like motif of ERM; the stretched complex structure under constant tensile forces <100 pN maintained on a stable quasi-equilibrium state, showing a high mechano-stabilization of the clamped complex; and, in consistent with the force-induced allostery at clamped stage, increasing tensile force (<50 pN) would decrease the complex dissociation probability but facilitate the phosphorylation site exposure, suggesting a force-enhanced biophysical connectivity of PSGL-1 signaling. These force-enhanced characters in both phosphorylation and unligation of ERM bound with PSGL-1 should be mediated by a catch-slip bond transition mechanism, in which four residue interactions on binding site were involved. This study might provide a novel insight into the transmembrane PSGL-1 signal, its biophysical connectivity and molecular structural basis for cellular immune responses in mechano-microenvironment, and showed a rational SMD-based computer strategy for predicting structure-function relation of protein under loads.


Asunto(s)
Proteínas del Citoesqueleto , Dominios FERM , Glicoproteínas de Membrana , Proteínas de la Membrana , Animales , Sitios de Unión , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Simulación de Dinámica Molecular , Unión Proteica
15.
Blood Adv ; 4(13): 3128-3135, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32649767

RESUMEN

Kindlin-3, a protein 4.1, ezrin, radixin, and moesin (FERM) domain-containing adaptor in hematopoietic cells, is essentially required for supporting the bidirectional integrin αIIbß3 signaling in platelets by binding to the integrin ß3 cytoplasmic tail. However, the structural details of kindlin-3's FERM domain remain unknown. In this study, we crystalized the kindlin-3's FERM domain protein and successfully solved its 3-dimensional structure. The structure shows that the 3 kindlin-3's FERM subdomains (F1, F2, and F3) compact together and form a cloverleaf-shaped conformation, which is stabilized by the binding interface between the F1 and F3 subdomains. Interestingly, the FERM domain of kindlin-3 exists as a monomer in both crystal and solution, which is different from its counterpart in kindlin-2 that is able to form a F2 subdomain-swapped dimer; nonetheless, dimerization is required for kindlin-3 to support integrin αIIbß3 activation, indicating that kindlin-3 may use alternative mechanisms for formation of a functional dimer in cells. To evaluate the functional importance of the cloverleaf-like FERM structure in kindlin-3, structure-based mutations were introduced into kindlin-3 to disrupt the F1/F3 interface. The results show that integrin αIIbß3 activation is significantly suppressed in platelets expressing the kindlin-3 mutant compared with those expressing wild-type kindlin-3. In addition, introduction of equivalent mutations into kindlin-1 and kindlin-2 also significantly compromises their ability to support integrin αIIbß3 activation in CHO cells. Together, our findings suggest that the cloverleaf-like FERM domain in kindlins is structurally important for supporting integrin αIIbß3 activation.


Asunto(s)
Plaquetas , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Animales , Cricetinae , Cricetulus , Dominios FERM , Integrina beta3 , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética
16.
J Biol Chem ; 295(39): 13570-13583, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32727844

RESUMEN

Hepatic abundance of the low-density lipoprotein receptor (LDLR) is a critical determinant of circulating plasma LDL cholesterol levels and hence development of coronary artery disease. The sterol-responsive E3 ubiquitin ligase inducible degrader of the LDLR (IDOL) specifically promotes ubiquitination and subsequent lysosomal degradation of the LDLR and thus controls cellular LDL uptake. IDOL contains an extended N-terminal FERM (4.1 protein, ezrin, radixin, and moesin) domain, responsible for substrate recognition and plasma membrane association, and a second C-terminal RING domain, responsible for the E3 ligase activity and homodimerization. As IDOL is a putative lipid-lowering drug target, we investigated the molecular details of its substrate recognition. We produced and isolated full-length IDOL protein, which displayed high autoubiquitination activity. However, in vitro ubiquitination of its substrate, the intracellular tail of the LDLR, was low. To investigate the structural basis for this, we determined crystal structures of the extended FERM domain of IDOL and multiple conformations of its F3ab subdomain. These reveal the archetypal F1-F2-F3 trilobed FERM domain structure but show that the F3c subdomain orientation obscures the target-binding site. To substantiate this finding, we analyzed the full-length FERM domain and a series of truncated FERM constructs by small-angle X-ray scattering (SAXS). The scattering data support a compact and globular core FERM domain with a more flexible and extended C-terminal region. This flexibility may explain the low activity in vitro and suggests that IDOL may require activation for recognition of the LDLR.


Asunto(s)
Receptores de LDL/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Sitios de Unión , Dominios FERM , Humanos , Modelos Moleculares , Receptores de LDL/química , Especificidad por Sustrato , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
17.
Chem Biol Drug Des ; 95(6): 584-599, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32080977

RESUMEN

The N-terminal FERM domain of focal adhesion kinase (FAK) contributes to FAK scaffolding and interacts with HER2, an oncogene and receptor tyrosine kinase. The interaction between HER2 and FAK drives resistance to FAK-kinase domain inhibitors through FAK Y397 transphosphorylation and FAK re-activation upon inhibition. As such, FAK FERM remains an attractive drug discovery target. In this report, we detail an alternative approach to targeting FAK through virtual screening-based discovery of chemical probes that target FAK FERM. We validated the binding interface between HER2 and FAK using site-directed mutagenesis and GST pull-down experiments. We assessed the ligandability of key-binding residues of HER2 and FAK utilizing computational tools. We developed a virtual screening method to screen ~200,000 compounds against the FAK FERM domain, identifying 20 virtual chemical probes. We performed GST pull-down screening on these compounds, discovering two hits, VS4 and VS14, with nanomolar IC50 s in disrupting HER2-FAK. We performed further testing, including molecular docking, immunofluorescence, phosphorylation, and cellular invasion assays to evaluate the compounds' biological effects. One probe, VS14, was identified with the ability to block both auto- and transphosphorylation of Y397. In all, these studies identify two new probes that target FAK FERM, enabling future investigation of this domain.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Secuencia de Aminoácidos , Dominios FERM , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Mutagénesis , Neoplasias , Fosforilación , Unión Proteica , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología
18.
Int J Oncol ; 55(5): 979-987, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31545421

RESUMEN

Protein 4.1B/DAL­1, encoded by erythrocyte membrane protein band 4.1­like 3 (EPB41L3), belongs to the protein 4.1 superfamily, a group of proteins that share a conserved four.one­ezrin­radixin­moesin (FERM) domain. Protein 4.1B/DAL­1 serves a crucial role in cytoskeletal organization and a number of processes through multiple interactions with membrane proteins via its FERM, spectrin­actin­binding and C­terminal domains. A number of studies have indicated that a loss of EPB41L3 expression is commonly observed in lung cancer, breast cancer, esophageal squamous cell carcinoma and meningiomas. DNA methylation and a loss of heterozygosity have been reported to contribute to the downregulation of EPB41L3. To date, the biological functions of protein 4.1B/DAL­1 in carcinogenesis remain unknown. The present review summarizes the current understanding of the role of protein 4.1B/DAL­1 in cancer and highlights its potential as a cancer diagnostic and prognostic biomarker in cancer therapeutics.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Microfilamentos/metabolismo , Neoplasias/patología , Dominios FERM , Humanos , Proteínas de Microfilamentos/genética , Neoplasias/genética , Neoplasias/metabolismo
19.
BMC Mol Cell Biol ; 20(1): 10, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31109284

RESUMEN

BACKGROUND: Focal Adhesion Kinase (FAK) is a major cancer drug target that is involved in numerous aspects of tumor progression and survival. While multiple research groups have developed ATP-competitive small molecule inhibitors that target the kinase enzyme, recent attention has been focused on the FAK FERM (Band 4.1, Ezrin, Radixin, Moesin) domain that contains key residue Y397 and contributes to many protein-protein interactions. Previous x-ray crystal structures of the FAK FERM domain gave conflicting results on the structure of the Y397 region and therefore the overall druggability. RESULTS: Here, we report the identification of a higher resolution crystal structure of the avian FAK FERM domain that shows conformational differences in Y397 and surrounding residues in the F1 lobe. In addition, we resolve the residues of the Src SH3 binding site, an area of the FERM domain that has previously shown limited electron density. CONCLUSIONS: These crystallographic data suggest that the Y397 region is highly dynamic and question the druggability of a putative pocket on the F1 lobe. In addition, new electron density data around the Src SH3 binding site provide structural insight on the FAK-Src activation cascade through a putative auto-inhibitory conformation.


Asunto(s)
Proteínas Aviares/química , Cristalización , Dominios FERM , Proteína-Tirosina Quinasas de Adhesión Focal/química , Proteínas Proto-Oncogénicas pp60(c-src)/química , Tirosina/química , Dominios Homologos src , Animales , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Proteínas del Citoesqueleto/química , Dimerización , Descubrimiento de Drogas/métodos , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas de Microfilamentos/química , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína
20.
Int J Pediatr Otorhinolaryngol ; 117: 115-126, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30579064

RESUMEN

OBJECTIVE: Hereditary hearing loss is the most common neurosensory disorder in humans caused by myriad mutations in numerous genes. Autosomal recessive nonsyndromic hearing loss (ARNSHL) accounts for 80% of hearing impairments of genetic origin and is quite prevalent in societies with a high rate of consanguinity. In the current study, we investigated the causes of sensorineural hearing loss in 24 unrelated Iranian families who were mainly consanguineous and had at least two affected children. METHODS: All probands were initially screened for GJB2 mutations, as the most common causes of ARNSHL in Iran. Verified GJB2-negative samples were subsequently subjected to whole exome sequencing (WES) to identify the underlying causes of hearing impairment, and the variants identified in each family were further confirmed by Sanger sequencing. RESULTS: WES revealed three previously unreported mutations in MYO15A, the gene encoding the unconventional myosin 15 (Myo15). All variants identified, c.C6436T (p.R2146W), c.C9584G (p.P3195R) and c.G10266C (p.Q3422H), reside in the MYTH4 (myosin tail homology) and FERM (4.1 ezrin, radixin, moesin) domains of the protein. CONCLUSION: Globally, mutations in MYO15A are considered to be among the most prevalent genetic causes of ARNSHL, and they rank as the third leading cause of hearing loss in the Iranian population, below GJB2 and SLC26A4. Yet again, these results endorse the importance of MYO15 screening in hearing impaired populations, particularly in Iran.


Asunto(s)
Sordera/genética , Dominios FERM/genética , Pérdida Auditiva Sensorineural/genética , Miosinas/genética , Consanguinidad , Femenino , Humanos , Irán , Masculino , Mutación , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...