Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Cell ; 174(4): 897-907.e14, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30078705

RESUMEN

Akt is a critical protein kinase that drives cancer proliferation, modulates metabolism, and is activated by C-terminal phosphorylation. The current structural model for Akt activation by C-terminal phosphorylation has centered on intramolecular interactions between the C-terminal tail and the N lobe of the kinase domain. Here, we employ expressed protein ligation to produce site-specifically phosphorylated forms of purified Akt1 that are well suited for mechanistic analysis. Using biochemical, crystallographic, and cellular approaches, we determine that pSer473-Akt activation is driven by an intramolecular interaction between the C-tail and the pleckstrin homology (PH)-kinase domain linker that relieves PH domain-mediated Akt1 autoinhibition. Moreover, dual phosphorylation at Ser477/Thr479 activates Akt1 through a different allosteric mechanism via an apparent activation loop interaction that reduces autoinhibition by the PH domain and weakens PIP3 affinity. These results provide a new framework for understanding how Akt is controlled in cell signaling and suggest distinct functions for differentially modified Akt forms.


Asunto(s)
Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo , Treonina/metabolismo , Cristalografía por Rayos X , Activación Enzimática , Células HCT116 , Humanos , Fosforilación , Dominios Homólogos a Pleckstrina , Unión Proteica , Conformación Proteica , Proteínas Proto-Oncogénicas c-akt/química , Serina/química , Transducción de Señal , Treonina/química
2.
Nature ; 628(8007): 416-423, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538786

RESUMEN

Antibody and chimeric antigen receptor (CAR) T cell-mediated targeted therapies have improved survival in patients with solid and haematologic malignancies1-9. Adults with T cell leukaemias and lymphomas, collectively called T cell cancers, have short survival10,11 and lack such targeted therapies. Thus, T cell cancers particularly warrant the development of CAR T cells and antibodies to improve patient outcomes. Preclinical studies showed that targeting T cell receptor ß-chain constant region 1 (TRBC1) can kill cancerous T cells while preserving sufficient healthy T cells to maintain immunity12, making TRBC1 an attractive target to treat T cell cancers. However, the first-in-human clinical trial of anti-TRBC1 CAR T cells reported a low response rate and unexplained loss of anti-TRBC1 CAR T cells13,14. Here we demonstrate that CAR T cells are lost due to killing by the patient's normal T cells, reducing their efficacy. To circumvent this issue, we developed an antibody-drug conjugate that could kill TRBC1+ cancer cells in vitro and cure human T cell cancers in mouse models. The anti-TRBC1 antibody-drug conjugate may provide an optimal format for TRBC1 targeting and produce superior responses in patients with T cell cancers.


Asunto(s)
Inmunoconjugados , Leucemia de Células T , Linfoma de Células T , Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T , Animales , Femenino , Humanos , Ratones , Inmunoconjugados/inmunología , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/inmunología , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 120(20): e2217096120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155902

RESUMEN

Diverse extracellular proteins negatively regulate WNT signaling. One such regulator is adenomatosis polyposis coli down-regulated 1 (APCDD1), a conserved single-span transmembrane protein. In response to WNT signaling in a variety of tissues, APCDD1 transcripts are highly up-regulated. We have determined the three-dimensional structure of the extracellular domain of APCDD1, and this structure reveals an unusual architecture consisting of two closely apposed ß-barrel domains (ABD1 and ABD2). ABD2, but not ABD1, has a large hydrophobic pocket that accommodates a bound lipid. The APCDD1 ECD can also bind to WNT7A, presumably via its covalently bound palmitoleate, a modification that is common to all WNTs and is essential for signaling. This work suggests that APCDD1 functions as a negative feedback regulator by titrating WNT ligands at the surface of responding cells.


Asunto(s)
Poliposis Adenomatosa del Colon , Proteínas de la Membrana , Humanos , Proteínas de la Membrana/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Vía de Señalización Wnt , Lípidos , beta Catenina/genética , beta Catenina/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(14): e2221103120, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36996108

RESUMEN

In many organs, small openings across capillary endothelial cells (ECs) allow the diffusion of low-molecular weight compounds and small proteins between the blood and tissue spaces. These openings contain a diaphragm composed of radially arranged fibers, and current evidence suggests that a single-span type II transmembrane protein, plasmalemma vesicle-associated protein-1 (PLVAP), constitutes these fibers. Here, we present the three-dimensional crystal structure of an 89-amino acid segment of the PLVAP extracellular domain (ECD) and show that it adopts a parallel dimeric alpha-helical coiled-coil configuration with five interchain disulfide bonds. The structure was solved using single-wavelength anomalous diffraction from sulfur-containing residues (sulfur SAD) to generate phase information. Biochemical and circular dichroism (CD) experiments show that a second PLVAP ECD segment also has a parallel dimeric alpha-helical configuration-presumably a coiled coil-held together with interchain disulfide bonds. Overall, ~2/3 of the ~390 amino acids within the PLVAP ECD adopt a helical configuration, as determined by CD. We also determined the sequence and epitope of MECA-32, an anti-PLVAP antibody. Taken together, these data lend strong support to the model of capillary diaphragms formulated by Tse and Stan in which approximately ten PLVAP dimers are arranged within each 60- to 80-nm-diameter opening like the spokes of a bicycle wheel. Passage of molecules through the wedge-shaped pores is presumably determined both by the length of PLVAP-i.e., the long dimension of the pore-and by the chemical properties of amino acid side chains and N-linked glycans on the solvent-accessible faces of PLVAP.


Asunto(s)
Diafragma , Células Endoteliales , Diafragma/metabolismo , Células Endoteliales/metabolismo , Proteínas Portadoras/metabolismo , Endotelio Vascular/metabolismo , Disulfuros/metabolismo , Dicroismo Circular
5.
J Biol Chem ; 300(3): 105715, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38309503

RESUMEN

NEDD4L is a HECT-type E3 ligase that catalyzes the addition of ubiquitin to intracellular substrates such as the cardiac voltage-gated sodium channel, NaV1.5. The intramolecular interactions of NEDD4L regulate its enzymatic activity which is essential for proteostasis. For NaV1.5, this process is critical as alterations in Na+ current is involved in cardiac diseases including arrhythmias and heart failure. In this study, we perform extensive biochemical and functional analyses that implicate the C2 domain and the first WW-linker (1,2-linker) in the autoregulatory mechanism of NEDD4L. Through in vitro and electrophysiological experiments, the NEDD4L 1,2-linker was determined to be important in substrate ubiquitination of NaV1.5. We establish the preferred sites of ubiquitination of NEDD4L to be in the second WW-linker (2,3-linker). Interestingly, NEDD4L ubiquitinates the cytoplasmic linker between the first and second transmembrane domains of the channel (DI-DII) of NaV1.5. Moreover, we design a genetically encoded modulator of Nav1.5 that achieves Na+ current reduction using the NEDD4L HECT domain as cargo of a NaV1.5-binding nanobody. These investigations elucidate the mechanisms regulating the NEDD4 family and furnish a new molecular framework for understanding NaV1.5 ubiquitination.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte , Canal de Sodio Activado por Voltaje NAV1.5 , Ubiquitina-Proteína Ligasas Nedd4 , Ubiquitinación , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Ubiquitina/metabolismo , Humanos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Células HEK293
6.
Mol Cell ; 66(3): 345-357.e6, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475870

RESUMEN

The HECT E3 ligases ubiquitinate numerous transcription factors and signaling molecules, and their activity must be tightly controlled to prevent cancer, immune disorders, and other diseases. In this study, we have found unexpectedly that peptide linkers tethering WW domains in several HECT family members are key regulatory elements of their catalytic activities. Biochemical, structural, and cellular analyses have revealed that the linkers can lock the HECT domain in an inactive conformation and block the proposed allosteric ubiquitin binding site. Such linker-mediated autoinhibition of the HECT domain can be relieved by linker post-translational modifications, but complete removal of the brake can induce hyperactive autoubiquitination and E3 self destruction. These results clarify the mechanisms of several HECT protein cancer associated mutations and provide a new framework for understanding how HECT ubiquitin ligases must be finely tuned to ensure normal cellular behavior.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación Alostérica , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Activación Enzimática , Estabilidad de Enzimas , Células HeLa , Humanos , Modelos Moleculares , Mutación , Ubiquitina-Proteína Ligasas Nedd4 , Fosforilación , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas Represoras/química , Proteínas Represoras/genética , Relación Estructura-Actividad , Transfección , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
7.
Proc Natl Acad Sci U S A ; 119(15): e2123406119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35394875

RESUMEN

HIV-1 infection is incurable due to the persistence of the virus in a latent reservoir of resting memory CD4+ T cells. "Shock-and-kill" approaches that seek to induce HIV-1 gene expression, protein production, and subsequent targeting by the host immune system have been unsuccessful due to a lack of effective latency-reversing agents (LRAs) and kill strategies. In an effort to develop reagents that could be used to promote killing of infected cells, we constructed T cell receptor (TCR)-mimic antibodies to HIV-1 peptide-major histocompatibility complexes (pMHC). Using phage display, we panned for phages expressing antibody-like variable sequences that bound HIV-1 pMHC generated using the common HLA-A*02:01 allele. We targeted three epitopes in Gag and reverse transcriptase identified and quantified via Poisson detection mass spectrometry from cells infected in vitro with a pseudotyped HIV-1 reporter virus (NL4.3 dEnv). Sequences isolated from phages that bound these pMHC were cloned into a single-chain diabody backbone (scDb) sequence, such that one fragment is specific for an HIV-1 pMHC and the other fragment binds to CD3ε, an essential signal transduction subunit of the TCR. Thus, these antibodies utilize the sensitivity of T cell signaling as readouts for antigen processing and as agents to promote killing of infected cells. Notably, these scDbs are exquisitely sensitive and specific for the peptide portion of the pMHC. Most importantly, one scDb caused killing of infected cells presenting a naturally processed target pMHC. This work lays the foundation for a novel therapeutic killing strategy toward elimination of the HIV-1 reservoir.


Asunto(s)
Anticuerpos Biespecíficos , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Linfocitos T CD4-Positivos , Humanos , Imitación Molecular , Receptores de Antígenos de Linfocitos T , Latencia del Virus
8.
Protein Expr Purif ; 215: 106406, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37995943

RESUMEN

The baculovirus expression system is a powerful and widely used method to generate large quantities of recombinant protein. However, challenges exist in workflows utilizing either liquid baculovirus stocks or the Titerless Infected-Cells Preservation and Scale-Up (TIPS) method, including the time and effort to generate baculoviruses, screen for protein expression and store large numbers of baculovirus stocks. To mitigate these challenges, we have developed a streamlined, hybrid workflow which utilizes high titer liquid virus stocks for rapid plate-based protein expression screening, followed by a TIPS-based scale-up for larger protein production efforts. Additionally, we have automated each step in this screening workflow using a custom robotic system. With these process improvements, we have significantly reduced the time, effort and resources required to manage large baculovirus generation and expression screening campaigns.


Asunto(s)
Baculoviridae , Triaje , Flujo de Trabajo , Baculoviridae/genética , Baculoviridae/metabolismo , Proteínas Recombinantes , Vectores Genéticos
9.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33731480

RESUMEN

Developing therapeutic agents with potent antitumor activity that spare normal tissues remains a significant challenge. Clonal loss of heterozygosity (LOH) is a widespread and irreversible genetic alteration that is exquisitely specific to cancer cells. We hypothesized that LOH events can be therapeutically targeted by "inverting" the loss of an allele in cancer cells into an activating signal. Here we describe a proof-of-concept approach utilizing engineered T cells approximating NOT-gate Boolean logic to target counterexpressed antigens resulting from LOH events in cancer. The NOT gate comprises a chimeric antigen receptor (CAR) targeting the allele of human leukocyte antigen (HLA) that is retained in the cancer cells and an inhibitory CAR (iCAR) targeting the HLA allele that is lost in the cancer cells. We demonstrate that engineered T cells incorporating such NOT-gate logic can be activated in a genetically predictable manner in vitro and in mice to kill relevant cancer cells. This therapeutic approach, termed NASCAR (Neoplasm-targeting Allele-Sensing CAR), could, in theory, be extended to LOH of other polymorphic genes that result in altered cell surface antigens in cancers.


Asunto(s)
Biomarcadores de Tumor , Inmunoterapia , Pérdida de Heterocigocidad , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/terapia , Alelos , Antígenos de Neoplasias/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva , Terapia Molecular Dirigida/efectos adversos , Terapia Molecular Dirigida/métodos , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/uso terapéutico
10.
J Biol Chem ; 298(5): 101854, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35331737

RESUMEN

WWP2 is a HECT E3 ligase that targets protein Lys residues for ubiquitination and is comprised of an N-terminal C2 domain, four central WW domains, and a C-terminal catalytic HECT domain. The peptide segment between the middle WW domains, the 2,3-linker, is known to autoinhibit the catalytic domain, and this autoinhibition can be relieved by phosphorylation at Tyr369. Several protein substrates of WWP2 have been identified, including the tumor suppressor lipid phosphatase PTEN, but the full substrate landscape and biological functions of WWP2 remain to be elucidated. Here, we used protein microarray technology and the activated enzyme phosphomimetic mutant WWP2Y369E to identify potential WWP2 substrates. We identified 31 substrate hits for WWP2Y369E using protein microarrays, of which three were known autophagy receptors (NDP52, OPTN, and SQSTM1). These three hits were validated with in vitro and cell-based transfection assays and the Lys ubiquitination sites on these proteins were mapped by mass spectrometry. Among the mapped ubiquitin sites on these autophagy receptors, many had been previously identified in the endogenous proteins. Finally, we observed that WWP2 KO SH-SH5Y neuroblastoma cells using CRISPR-Cas9 showed a defect in mitophagy, which could be rescued by WWP2Y369E transfection. These studies suggest that WWP2-mediated ubiquitination of the autophagy receptors NDP52, OPTN, and SQSTM1 may positively contribute to the regulation of autophagy.


Asunto(s)
Autofagia , Análisis por Matrices de Proteínas , Ubiquitina-Proteína Ligasas , Proteínas de Ciclo Celular/metabolismo , Humanos , Proteínas de Transporte de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Proteína Sequestosoma-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
11.
J Biol Chem ; 298(4): 101763, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35202650

RESUMEN

Voltage-gated sodium channels, NaVs, are responsible for the rapid rise of action potentials in excitable tissues. NaV channel mutations have been implicated in several human genetic diseases, such as hypokalemic periodic paralysis, myotonia, and long-QT and Brugada syndromes. Here, we generated high-affinity anti-NaV nanobodies (Nbs), Nb17 and Nb82, that recognize the NaV1.4 (skeletal muscle) and NaV1.5 (cardiac muscle) channel isoforms. These Nbs were raised in llama (Lama glama) and selected from a phage display library for high affinity to the C-terminal (CT) region of NaV1.4. The Nbs were expressed in Escherichia coli, purified, and biophysically characterized. Development of high-affinity Nbs specifically targeting a given human NaV isoform has been challenging because they usually show undesired crossreactivity for different NaV isoforms. Our results show, however, that Nb17 and Nb82 recognize the CTNaV1.4 or CTNaV1.5 over other CTNav isoforms. Kinetic experiments by biolayer interferometry determined that Nb17 and Nb82 bind to the CTNaV1.4 and CTNaV1.5 with high affinity (KD ∼ 40-60 nM). In addition, as proof of concept, we show that Nb82 could detect NaV1.4 and NaV1.5 channels in mammalian cells and tissues by Western blot. Furthermore, human embryonic kidney cells expressing holo NaV1.5 channels demonstrated a robust FRET-binding efficiency for Nb17 and Nb82. Our work lays the foundation for developing Nbs as anti-NaV reagents to capture NaVs from cell lysates and as molecular visualization agents for NaVs.


Asunto(s)
Anticuerpos de Dominio Único , Canales de Sodio Activados por Voltaje , Animales , Células Cultivadas , Escherichia coli/genética , Humanos , Síndrome de QT Prolongado/metabolismo , Mamíferos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/metabolismo , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo
12.
Curr Top Microbiol Immunol ; 436: 3-49, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36243838

RESUMEN

This chapter is an introduction to phosphoinositide 3-kinases (PI3K), with class I PI3Ks as the central focus. First, the various PI3K isoforms in class I are presented with emphasis on their overall structure, subunits, subunit constitutive domains, domain-domain interactions, and functional relevance. This structural analysis is followed by a comprehensive history of seminal investigations into PI3K activity. Next, we highlight the divergent roles of the isoforms: PI3Kα, PI3Kß, PI3Kδ, and PI3Kγ. This section details signaling pathways in which these PI3K isoforms are involved, including the key upstream regulators of PI3K activity and some downstream cellular effects. Nodes of the PI3K pathway are also presented. Inhibitors of some isoforms are discussed to give an overview of the basis of some immunotherapies that are being used to target cell signaling. Finally, the chapter ends with a discussion of the dysregulation of PI3Ks in diseases including APDS, asthma, arthritis, and oncogenic mutations.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Transducción de Señal , Biología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Transducción de Señal/fisiología
13.
Proc Natl Acad Sci U S A ; 117(26): 15104-15111, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32541044

RESUMEN

Five small protein domains, the CC-domains, at the N terminus of the RECK protein, play essential roles in signaling by WNT7A and WNT7B in the context of central nervous system angiogenesis and blood-brain barrier formation and maintenance. We have determined the structure of CC domain 4 (CC4) at 1.65-Å resolution and find that it folds into a compact four-helix bundle with three disulfide bonds. The CC4 structure, together with homology modeling of CC1, reveals the surface locations of critical residues that were shown in previous mutagenesis studies to mediate GPR124 binding and WNT7A/WNT7B recognition and signaling. Surprisingly, sequence and structural homology searches reveal no other cell-surface or secreted domains in vertebrates that resemble the CC domain, a pattern that is in striking contrast to other ancient and similarly sized domains, such as Epidermal Growth Factor, Fibronectin Type 3, Immunoglobulin, and Thrombospondin type 1 domains, which are collectively present in hundreds of proteins.


Asunto(s)
Evolución Molecular , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans , Proteínas Ligadas a GPI/genética , Humanos , Ratones , Dominios Proteicos , Alineación de Secuencia
14.
J Biol Chem ; 296: 100832, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34048713

RESUMEN

Voltage-gated sodium channels (NaVs) underlie the initiation of action potentials in various excitable cell types and are regulated by channel-interacting proteins, including the cellular calcium sensor calmodulin and fibroblast growth factor homologous factors. Both of these are known to bind the NaV cytosolic C-terminal domain and modulate the channel's electrophysiology, but it was unknown whether they had any allosteric interactions with each other. A recent rigorous study provides insights into the molecular interactions of these ion channels and their partners that crucially take the cellular landscape into consideration.


Asunto(s)
Canales de Sodio Activados por Voltaje/fisiología , Animales , Humanos
15.
Proc Natl Acad Sci U S A ; 115(13): E3026-E3035, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29531055

RESUMEN

Calmodulin (CaM) regulation of voltage-gated calcium (CaV) channels is a powerful Ca2+ feedback mechanism that adjusts Ca2+ influx, affording rich mechanistic insights into Ca2+ decoding. CaM possesses a dual-lobed architecture, a salient feature of the myriad Ca2+-sensing proteins, where two homologous lobes that recognize similar targets hint at redundant signaling mechanisms. Here, by tethering CaM lobes, we demonstrate that bilobal architecture is obligatory for signaling to CaV channels. With one lobe bound, CaV carboxy tail rearranges itself, resulting in a preinhibited configuration precluded from Ca2+ feedback. Reconstitution of two lobes, even as separate molecules, relieves preinhibition and restores Ca2+ feedback. CaV channels thus detect the coincident binding of two Ca2+-free lobes to promote channel opening, a molecular implementation of a logical NOR operation that processes spatiotemporal Ca2+ signals bifurcated by CaM lobes. Overall, a unified scheme of CaV channel regulation by CaM now emerges, and our findings highlight the versatility of CaM to perform exquisite Ca2+ computations.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Calmodulina/metabolismo , Activación del Canal Iónico/fisiología , Secuencia de Aminoácidos , Animales , Canales de Calcio/química , Calmodulina/química , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Ratas , Homología de Secuencia , Transducción de Señal
16.
Biochemistry ; 59(29): 2751-2759, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32584028

RESUMEN

Farnesyl diphosphate synthase (FPPS) is an isoprenoid chain elongation enzyme that catalyzes the sequential condensation of dimethylallyl diphosphate (C5) with isopentenyl diphosphate (IPP; C5) and the resulting geranyl diphosphate (GPP; C10) with another molecule of IPP, eventually producing farnesyl diphosphate (FPP; C15), which is a precursor for the biosynthesis of a vast majority of isoprenoids. Previous studies of FPPS have highlighted the importance of the structure around the hydrophobic chain elongation path in determining product specificity. To investigate what structural features define the final chain length of the product in FPPS from Leishmania major, we designed and expressed six mutants of LmFPPS by replacing small amino acids around the binding pocket with bulky residues. Using enzymatic assays, binding kinetics, and crystallographic studies, we analyzed the effects of these mutations on the activity and product specificity of FPPS. Our results revealed that replacement of Thr-164 with tryptophan and phenylalanine completely abolished the activity of FPPS. Intriguingly, the T164Y substitution displayed dual product specificity and produced a mixture GPP and FPP as final products, with an activity for FPP synthesis that was lower than that of the wild-type enzyme. These data indicate that Thr-164 is a potential regulator of product specificity.


Asunto(s)
Geraniltranstransferasa/metabolismo , Leishmania major/enzimología , Sitios de Unión , Cristalografía por Rayos X , Difosfatos/metabolismo , Diterpenos/metabolismo , Geraniltranstransferasa/química , Hemiterpenos/metabolismo , Humanos , Leishmania major/química , Leishmania major/metabolismo , Leishmaniasis Cutánea/parasitología , Modelos Moleculares , Compuestos Organofosforados/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Conformación Proteica , Sesquiterpenos/metabolismo , Especificidad por Sustrato
17.
J Biol Chem ; 294(50): 19322-19334, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31690625

RESUMEN

Mutations in CTNNB1, the gene encoding ß-catenin, are common in colon and liver cancers, the most frequent mutation affecting Ser-45 in ß-catenin. Peptides derived from WT ß-catenin have previously been shown to be presented on the cell surface as part of major histocompatibility complex (MHC) class I, suggesting an opportunity for targeting this common driver gene mutation with antibody-based therapies. Here, crystal structures of both the WT and S45F mutant peptide bound to HLA-A*03:01 at 2.20 and 2.45 Å resolutions, respectively, confirmed the accessibility of the phenylalanine residue for antibody recognition. Phage display was then used to identify single-chain variable fragment clones that selectively bind the S45F mutant peptide presented in HLA-A*03:01 and have minimal WT or other off-target binding. Following the initial characterization of five clones, we selected a single clone, E10, for further investigation. We developed a computational model of the binding of E10 to the mutant peptide-bound HLA-A3, incorporating data from affinity maturation as initial validation. In the future, our model may be used to design clones with maintained specificity and higher affinity. Such derivatives could be adapted into either cell-based (CAR-T) or protein-based (bispecific T-cell engagers) therapies to target cancer cells harboring the S45F mutation in CTNNB1.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/genética , Fragmentos de Inmunoglobulinas/química , Ingeniería de Proteínas , beta Catenina/genética , Línea Celular , Antígenos de Histocompatibilidad Clase I/química , Humanos , Modelos Moleculares , Mutación , beta Catenina/química
18.
Molecules ; 25(11)2020 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-32503272

RESUMEN

Neglected tropical diseases such as Chagas disease and leishmaniasis affect millions of people around the world. Both diseases affect various parts of the globe and drugs traditionally used in therapy against these diseases have limitations, especially with regard to low efficacy and high toxicity. In this context, the class of bisphosphonate-based compounds has made significant advances regarding the chemical synthesis process as well as the pharmacological properties attributed to these compounds. Among this spectrum of pharmacological activity, bisphosphonate compounds with antiparasitic activity stand out, especially in the treatment of Chagas disease and leishmaniasis caused by Trypanosoma cruzi and Leishmania spp., respectively. Some bisphosphonate compounds can inhibit the mevalonate pathway, an essential metabolic pathway, by interfering with the synthesis of ergosterol, a sterol responsible for the growth and viability of these parasites. Therefore, this review aims to present the information about the importance of these compounds as antiparasitic agents and as potential new drugs to treat Chagas disease and leishmaniasis.


Asunto(s)
Enfermedad de Chagas/tratamiento farmacológico , Difosfonatos/farmacología , Leishmania infantum/efectos de los fármacos , Leishmaniasis/tratamiento farmacológico , Trypanosoma cruzi/efectos de los fármacos , Animales , Antiparasitarios , Enfermedad de Chagas/parasitología , Humanos , Leishmaniasis/parasitología
19.
J Biol Chem ; 293(11): 4134-4148, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29378847

RESUMEN

CoA is the major acyl carrier in mammals and a key cofactor in energy metabolism. Dynamic regulation of CoA in different tissues and organs supports metabolic flexibility. Two mammalian Nudix hydrolases, Nudt19 and Nudt7, degrade CoA in vitro Nudt19 and Nudt7 possess conserved Nudix and CoA signature sequences and specifically hydrolyze the diphosphate bond of free CoA and acyl-CoAs to form 3',5'-ADP and 4'-(acyl)phosphopantetheine. Limited information is available on these enzymes, but the relatively high abundance of Nudt19 and Nudt7 mRNA in the kidney and liver, respectively, suggests that they play specific roles in the regulation of CoA levels in these organs. Here, we analyzed Nudt19-/- mice and found that deletion of Nudt19 elevates kidney CoA levels in mice fed ad libitum, indicating that Nudt19 contributes to the regulation of CoA in vivo Unlike what was observed for the regulation of Nudt7 in the liver, Nudt19 transcript and protein levels in the kidney did not differ between fed and fasted states. Instead, we identified chenodeoxycholic acid as a specific Nudt19 inhibitor that competed with CoA for Nudt19 binding but did not bind to Nudt7. Exchange of the Nudix and CoA signature motifs between the two isoforms dramatically decreased their kcat Furthermore, substitutions of conserved residues within these motifs identified amino acids playing different roles in CoA binding and hydrolysis in Nudt19 and Nudt7. Our results reveal that the kidney and liver each possesses a distinct peroxisomal CoA diphosphohydrolase.


Asunto(s)
Coenzima A/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Pirofosfatasas/fisiología , Secuencia de Aminoácidos , Animales , Isoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación Proteica , Pirofosfatasas/química , Homología de Secuencia , Hidrolasas Nudix
20.
Nat Mater ; 17(1): 79-89, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29115293

RESUMEN

Some protein components of intracellular non-membrane-bound entities, such as RNA granules, are known to form hydrogels in vitro. The physico-chemical properties and functional role of these intracellular hydrogels are difficult to study, primarily due to technical challenges in probing these materials in situ. Here, we present iPOLYMER, a strategy for a rapid induction of protein-based hydrogels inside living cells that explores the chemically inducible dimerization paradigm. Biochemical and biophysical characterizations aided by computational modelling show that the polymer network formed in the cytosol resembles a physiological hydrogel-like entity that acts as a size-dependent molecular sieve. We functionalize these polymers with RNA-binding motifs that sequester polyadenine-containing nucleotides to synthetically mimic RNA granules. These results show that iPOLYMER can be used to synthetically reconstitute the nucleation of biologically functional entities, including RNA granules in intact cells.


Asunto(s)
Hidrogeles/metabolismo , Polímeros/metabolismo , ARN/metabolismo , Animales , Materiales Biocompatibles , Células COS , Chlorocebus aethiops
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA