Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 133(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37317963

RESUMEN

RAS mutations are among the most prevalent oncogenic drivers in cancers. RAS proteins propagate signals only when associated with cellular membranes as a consequence of lipid modifications that impact their trafficking. Here, we discovered that RAB27B, a RAB family small GTPase, controlled NRAS palmitoylation and trafficking to the plasma membrane, a localization required for activation. Our proteomic studies revealed RAB27B upregulation in CBL- or JAK2-mutated myeloid malignancies, and its expression correlated with poor prognosis in acute myeloid leukemias (AMLs). RAB27B depletion inhibited the growth of CBL-deficient or NRAS-mutant cell lines. Strikingly, Rab27b deficiency in mice abrogated mutant but not WT NRAS-mediated progenitor cell growth, ERK signaling, and NRAS palmitoylation. Further, Rab27b deficiency significantly reduced myelomonocytic leukemia development in vivo. Mechanistically, RAB27B interacted with ZDHHC9, a palmitoyl acyltransferase that modifies NRAS. By regulating palmitoylation, RAB27B controlled c-RAF/MEK/ERK signaling and affected leukemia development. Importantly, RAB27B depletion in primary human AMLs inhibited oncogenic NRAS signaling and leukemic growth. We further revealed a significant correlation between RAB27B expression and sensitivity to MEK inhibitors in AMLs. Thus, our studies presented a link between RAB proteins and fundamental aspects of RAS posttranslational modification and trafficking, highlighting future therapeutic strategies for RAS-driven cancers.


Asunto(s)
Leucemia Mieloide , Lipoilación , Humanos , Animales , Ratones , Proteómica , Transducción de Señal , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas de la Membrana/genética , GTP Fosfohidrolasas
2.
Biochem Soc Trans ; 51(3): 1191-1199, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37222266

RESUMEN

RAS proteins are small GTPases that transduce signals from membrane receptors to signaling pathways that regulate growth and differentiation. Four RAS proteins are encoded by three genes - HRAS, KRAS, NRAS. Among them, KRAS is mutated in human cancer more frequently than any other oncogene. The KRAS pre-mRNA is alternatively spliced to generate two transcripts, KRAS4A and KRAS4B, that encode distinct proto-oncoproteins that differ almost exclusively in their C-terminal hypervariable regions (HVRs) that controls subcellular trafficking and membrane association. The KRAS4A isoform arose 475 million years ago in jawed vertebrates and has persisted in all vertebrates ever since, strongly suggesting non-overlapping functions of the splice variants. Because KRAS4B is expressed at higher levels in most tissues, it has been considered the principal KRAS isoform. However, emerging evidence for KRAS4A expression in tumors and splice variant-specific interactions and functions have sparked interest in this gene product. Among these findings, the KRAS4A-specific regulation of hexokinase I is a stark example. The aim of this mini-review is to provide an overview of the origin and differential functions of the two splice variants of KRAS.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Animales , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Mutación
3.
Oncogene ; 42(21): 1741-1750, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37031342

RESUMEN

KRAS, HRAS and NRAS proto-oncogenes belong to a family of 40 highly homologous genes, which in turn are a subset of a superfamily of >160 genes encoding small GTPases. RAS proteins consist of a globular G-domain (aa1-166) and a 22-23 aa unstructured hypervariable region (HVR) that mediates membrane targeting. The evolutionary origins of the RAS isoforms, their HVRs and alternative splicing of the KRAS locus has not been explored. We found that KRAS is basal to the RAS proto-oncogene family and its duplication generated HRAS in the common ancestor of vertebrates. In a second round of duplication HRAS generated NRAS and KRAS generated an additional RAS gene we have designated KRASBL, absent in mammals and birds. KRAS4A arose through a duplication and insertion of the 4th exon of NRAS into the 3rd intron of KRAS. We found evolutionary conservation of a short polybasic region (PBR1) in HRAS, NRAS and KRAS4A, a second polybasic region (PBR2) in KRAS4A, two neutralized basic residues (NB) and a serine in KRAS4B and KRASBL, and a modification of the CaaX motif in vertebrates with farnesyl rather than geranylgeranyl polyisoprene lipids, suggesting that a less hydrophobic membrane anchor is critical to RAS protein function. The persistence of four RAS isoforms through >400 million years of evolution argues strongly for differential function.


Asunto(s)
Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras , Animales , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Isoformas de Proteínas/genética , Proteínas ras/genética , Proteínas ras/metabolismo , Genes ras , Mamíferos/genética
4.
Mol Cell ; 83(8): 1210-1215, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36990093

RESUMEN

One of the open questions in RAS biology is the existence of RAS dimers and their role in RAF dimerization and activation. The idea of RAS dimers arose from the discovery that RAF kinases function as obligate dimers, which generated the hypothesis that RAF dimer formation might be nucleated by G-domain-mediated RAS dimerization. Here, we review the evidence for RAS dimerization and describe a recent discussion among RAS researchers that led to a consensus that the clustering of two or more RAS proteins is not due to the stable association of G-domains but, instead, is a consequence of RAS C-terminal membrane anchors and the membrane phospholipids with which they interact.


Asunto(s)
Quinasas raf , Proteínas ras , Dimerización , Consenso , Proteínas ras/genética , Proteínas ras/metabolismo , Quinasas raf/genética , Quinasas raf/metabolismo , Lípidos , Proteínas Proto-Oncogénicas c-raf/metabolismo
5.
Res Sq ; 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36711820

RESUMEN

KRAS, HRAS and NRAS oncogenes belong to a family of 40 highly homologous genes, which in turn are a subset of a superfamily of >160 genes encoding small GTPases. RAS oncoproteins consist of a globular G-domain (aa1-166) and a 22-23aa unstructured hypervariable region (HVR) that mediates membrane targeting. The evolutionarily origins of the RAS isoforms, their HVRs and alternative splicing of the KRAS locus has not been explored. We found that KRAS is basal to the oncogene family and its duplication generated HRAS in the common ancestor of vertebrates. In a second round of duplication HRAS generated NRAS and KRAS generated an additional RAS gene we have designated KRASBL, absent in mammals and birds. KRAS4A arose through a duplication and insertion of the 4th exon of NRAS into the 3rd intron of KRAS. We found evolutionarily conservation of a short polybasic region (PBR1) in HRAS, NRAS and KRAS4A, a second polybasic region (PBR2) in KRAS4A, two neutralized basic residues (NB) and a serine in KRAS4B and KRASBL, and a modification of the CaaX motif in vertebrates with farnesyl rather than geranylgeranyl polyisoprene lipids, suggesting that a less hydrophobic membrane anchor is critical to RAS oncoprotein function. The persistence of four RAS isoforms through >400 MY of evolution argues strongly for differential function.

6.
Front Cell Dev Biol ; 10: 1033348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36393833

RESUMEN

The three mammalian RAS genes (HRAS, NRAS and KRAS) encode four proteins that play central roles in cancer biology. Among them, KRAS is mutated more frequently in human cancer than any other oncogene. The pre-mRNA of KRAS is alternatively spliced to give rise to two products, KRAS4A and KRAS4B, which differ in the membrane targeting sequences at their respective C-termini. Notably, both KRAS4A and KRAS4B are oncogenic when KRAS is constitutively activated by mutation in exon 2 or 3. Whereas KRAS4B is the most studied oncoprotein, KRAS4A is understudied and until recently considered relatively unimportant. Emerging work has confirmed expression of KRAS4A in cancer and found non-overlapping functions of the splice variants. The most clearly demonstrated of these is direct regulation of hexokinase 1 by KRAS4A, suggesting that the metabolic vulnerabilities of KRAS-mutant tumors may be determined in part by the relative expression of the splice variants. The aim of this review is to address the most relevant characteristics and differential functions of the KRAS splice variants as they relate to cancer onset and progression.

7.
Curr Opin Struct Biol ; 71: 180-192, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34365229

RESUMEN

Mutations of RAS genes drive cancer more frequently than any other oncogene. RAS proteins integrate signals from a wide array of receptors and initiate downstream signaling through pathways that control cellular growth. RAS proteins are fundamentally binary molecular switches in which the off/on state is determined by the binding of GDP or GTP, respectively. As such, the intrinsic and regulated nucleotide-binding and hydrolytic properties of the RAS GTPase were historically believed to account for the entirety of the regulation of RAS signaling. However, it is increasingly clear that RAS proteins are also regulated by a vast array of post-translational modifications (PTMs). The current challenge is to understand what are the functional consequences of these modifications and which are physiologically relevant. Because PTMs are catalyzed by enzymes that may offer targets for drug discovery, the study of RAS PTMs has been a high priority for RAS biologists.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteínas ras , Proteínas Portadoras , Transducción de Señal , Proteínas ras/metabolismo
8.
Nat Commun ; 12(1): 4905, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385458

RESUMEN

α-ketoglutarate (KG), also referred to as 2-oxoglutarate, is a key intermediate of cellular metabolism with pleiotropic functions. Cell-permeable esterified analogs are widely used to study how KG fuels bioenergetic and amino acid metabolism and DNA, RNA, and protein hydroxylation reactions, as cellular membranes are thought to be impermeable to KG. Here we show that esterified KG analogs rapidly hydrolyze in aqueous media, yielding KG that, in contrast to prevailing assumptions, imports into many cell lines. Esterified KG analogs exhibit spurious KG-independent effects on cellular metabolism, including extracellular acidification, arising from rapid hydrolysis and de-protonation of α-ketoesters, and significant analog-specific inhibitory effects on glycolysis or mitochondrial respiration. We observe that imported KG decarboxylates to succinate in the cytosol and contributes minimally to mitochondrial metabolism in many cell lines cultured in normal conditions. These findings demonstrate that nuclear and cytosolic KG-dependent reactions may derive KG from functionally distinct subcellular pools and sources.


Asunto(s)
Aminoácidos/metabolismo , Metabolismo Energético , Ésteres/metabolismo , Ácidos Cetoglutáricos/metabolismo , Mitocondrias/metabolismo , Ácido Succínico/metabolismo , Animales , Línea Celular Tumoral , Citosol/metabolismo , Ésteres/química , Glucólisis , Células HEK293 , Humanos , Hidrólisis , Interacciones Hidrofóbicas e Hidrofílicas , Ácidos Cetoglutáricos/química , Ratones , Consumo de Oxígeno , Células RAW 264.7
9.
Elife ; 102021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33526168

RESUMEN

A farnesylated and methylated form of prelamin A called progerin causes Hutchinson-Gilford progeria syndrome (HGPS). Inhibiting progerin methylation by inactivating the isoprenylcysteine carboxylmethyltransferase (ICMT) gene stimulates proliferation of HGPS cells and improves survival of Zmpste24-deficient mice. However, we don't know whether Icmt inactivation improves phenotypes in an authentic HGPS mouse model. Moreover, it is unknown whether pharmacologic targeting of ICMT would be tolerated by cells and produce similar cellular effects as genetic inactivation. Here, we show that knockout of Icmt improves survival of HGPS mice and restores vascular smooth muscle cell numbers in the aorta. We also synthesized a potent ICMT inhibitor called C75 and found that it delays senescence and stimulates proliferation of late-passage HGPS cells and Zmpste24-deficient mouse fibroblasts. Importantly, C75 did not influence proliferation of wild-type human cells or Zmpste24-deficient mouse cells lacking Icmt, indicating drug specificity. These results raise hopes that ICMT inhibitors could be useful for treating children with HGPS.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Progeria/tratamiento farmacológico , Proteína Metiltransferasas/efectos de los fármacos , Piranos/farmacología , Animales , Aorta/patología , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Lamina Tipo A/metabolismo , Ratones , Ratones Noqueados , Miocitos del Músculo Liso , Progeria/genética , Progeria/patología , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/metabolismo
10.
Life Sci Alliance ; 4(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33579760

RESUMEN

Isoprenylcysteine carboxyl methyltransferase (ICMT) is the third of three enzymes that sequentially modify the C-terminus of CaaX proteins, including RAS. Although all four RAS proteins are substrates for ICMT, each traffics to membranes differently by virtue of their hypervariable regions that are differentially palmitoylated. We found that among RAS proteins, NRAS was unique in requiring ICMT for delivery to the PM, a consequence of having only a single palmitoylation site as its secondary affinity module. Although not absolutely required for palmitoylation, acylation was diminished in the absence of ICMT. Photoactivation and FRAP of GFP-NRAS revealed increase flux at the Golgi, independent of palmitoylation, in the absence of ICMT. Association of NRAS with the prenyl-protein chaperone PDE6δ also required ICMT and promoted anterograde trafficking from the Golgi. We conclude that carboxyl methylation of NRAS is required for efficient palmitoylation, PDE6δ binding, and homeostatic flux through the Golgi, processes that direct delivery to the plasma membrane.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Metiltransferasas/metabolismo , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/fisiología , GTP Fosfohidrolasas/fisiología , Humanos , Lipoilación/fisiología , Proteínas de la Membrana/fisiología , Proteína Metiltransferasas/fisiología , Transporte de Proteínas/fisiología , Proteínas ras
11.
Acad Med ; 96(4): 518-521, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464738

RESUMEN

PROBLEM: Physician-scientists are individuals trained in both clinical practice and scientific research. Often, the goal of physician-scientist training is to address pressing questions in biomedical research. The established pathways to formally train such individuals are mainly MD-PhD programs and physician-scientist track residencies. Although graduates of these pathways are well equipped to be physician-scientists, numerous factors, including funding and length of training, discourage application to such programs and impede success rates. APPROACH: To address some of the pressing challenges in training and retaining burgeoning physician-scientists, New York University Grossman School of Medicine formed the Accelerated MD-PhD-Residency Pathway in 2016. This pathway builds on the previously established accelerated 3-year MD pathway to residency at the same institution. The Accelerated MD-PhD-Residency Pathway conditionally accepts MD-PhD trainees to a residency position at the same institution through the National Resident Matching Program. OUTCOMES: Since its inception, 2 students have joined the Accelerated MD-PhD-Residency Pathway, which provides protected research time in their chosen residency. The pathway reduces the time to earn an MD and PhD by 1 year and reduces the MD training phase to 3 years, reducing the cost and lowering socioeconomic barriers. Remaining at the same institution for residency allows for the growth of strong research collaborations and mentoring opportunities, which foster success. NEXT STEPS: The authors and institutional leaders plan to increase the number of trainees who are accepted into the Accelerated MD-PhD-Residency Pathway and track the success of these students through residency and into practice to determine if the pathway is meeting its goal of increasing the number of practicing physician-scientists. The authors hope this model can serve as an example to leaders at other institutions who may wish to adopt this pathway for the training of their MD-PhD students.


Asunto(s)
Investigación Biomédica/educación , Investigación Biomédica/tendencias , Educación de Postgrado en Medicina/normas , Educación de Postgrado en Medicina/tendencias , Guías como Asunto , Internado y Residencia/normas , Internado y Residencia/tendencias , Adulto , Investigación Biomédica/estadística & datos numéricos , Educación de Postgrado en Medicina/estadística & datos numéricos , Femenino , Predicción , Humanos , Internado y Residencia/estadística & datos numéricos , Masculino , New York , Adulto Joven
12.
Proc Natl Acad Sci U S A ; 117(50): 31914-31922, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257571

RESUMEN

Inhibiting membrane association of RAS has long been considered a rational approach to anticancer therapy, which led to the development of farnesyltransferase inhibitors (FTIs). However, FTIs proved ineffective against KRAS-driven tumors. To reveal alternative therapeutic strategies, we carried out a genome-wide CRISPR-Cas9 screen designed to identify genes required for KRAS4B membrane association. We identified five enzymes in the prenylation pathway and SAFB, a nuclear protein with both DNA and RNA binding domains. Silencing SAFB led to marked mislocalization of all RAS isoforms as well as RAP1A but not RAB7A, a pattern that phenocopied silencing FNTA, the prenyltransferase α subunit shared by farnesyltransferase and geranylgeranyltransferase type I. We found that SAFB promoted RAS membrane association by controlling FNTA expression. SAFB knockdown decreased GTP loading of RAS, abrogated alternative prenylation, and sensitized RAS-mutant cells to growth inhibition by FTI. Our work establishes the prenylation pathway as paramount in KRAS membrane association, reveals a regulator of prenyltransferase expression, and suggests that reduction in FNTA expression may enhance the efficacy of FTIs.


Asunto(s)
Membrana Celular/metabolismo , Dimetilaliltranstransferasa/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Neoplasias/patología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores de Estrógenos/metabolismo , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Sistemas CRISPR-Cas/genética , Biología Computacional , Conjuntos de Datos como Asunto , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Neoplasias/genética , Proteínas Asociadas a Matriz Nuclear/genética , Prenilación de Proteína , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Estrógenos/genética
13.
Clin Immunol ; 217: 108485, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32504780

RESUMEN

T cell co-stimulation is important for the maintenance of immunologic tolerance. Co-inhibitory receptors including programmed cell death-1 (PD-1) confer peripheral tolerance to prevent autoimmunity. SAP (SH2D1A) is an adaptor molecule that is important in T cell signaling and has been shown to interact with signaling lymphocytic activation molecule (SLAM) family receptors also in the context of self-tolerance. We recently reported that SAP interferes with PD-1 function. In the current study, we investigated the levels of SAP and PD-1 in patients with rheumatoid arthritis (RA) to further understand what role they play in disease activity. We observed increased SAP levels in lymphocytes of RA patients and found that PD-1 levels correlated positively with RA disease activity. Additionally, we found that SAP interacts with CD28 to inhibit T cell signaling in vitro. This work demonstrates a putative molecular mechanism for SAP mediated PD-1 inhibition.


Asunto(s)
Artritis Reumatoide/inmunología , Antígenos CD28/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/metabolismo , Linfocitos T/inmunología , Adulto , Anciano , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Autoinmunidad/genética , Autoinmunidad/fisiología , Línea Celular Tumoral , Femenino , Humanos , Células Jurkat , Masculino , Persona de Mediana Edad , Fosforilación , Receptor de Muerte Celular Programada 1/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/genética , Adulto Joven
14.
Cancer Discov ; 10(7): 1018-1037, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32341021

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) evolves a complex microenvironment comprised of multiple cell types, including pancreatic stellate cells (PSC). Previous studies have demonstrated that stromal supply of alanine, lipids, and nucleotides supports the metabolism, growth, and therapeutic resistance of PDAC. Here we demonstrate that alanine cross-talk between PSCs and PDAC is orchestrated by the utilization of specific transporters. PSCs utilize SLC1A4 and other transporters to rapidly exchange and maintain environmental alanine concentrations. Moreover, PDAC cells upregulate SLC38A2 to supply their increased alanine demand. Cells lacking SLC38A2 fail to concentrate intracellular alanine and undergo a profound metabolic crisis resulting in markedly impaired tumor growth. Our results demonstrate that stromal-cancer metabolic niches can form through differential transporter expression, creating unique therapeutic opportunities to target metabolic demands of cancer. SIGNIFICANCE: This work identifies critical neutral amino acid transporters involved in channeling alanine between pancreatic stellate and PDAC cells. Targeting PDAC-specific alanine uptake results in a metabolic crisis impairing metabolism, proliferation, and tumor growth. PDAC cells specifically activate and require SLC38A2 to fuel their alanine demands that may be exploited therapeutically.This article is highlighted in the In This Issue feature, p. 890.


Asunto(s)
Adenocarcinoma/fisiopatología , Alanina/metabolismo , Carcinoma Ductal Pancreático/fisiopatología , Humanos , Redes y Vías Metabólicas , Transducción de Señal , Microambiente Tumoral
15.
Nature ; 576(7787): 482-486, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31827279

RESUMEN

The most frequently mutated oncogene in cancer is KRAS, which uses alternative fourth exons to generate two gene products (KRAS4A and KRAS4B) that differ only in their C-terminal membrane-targeting region1. Because oncogenic mutations occur in exons 2 or 3, two constitutively active KRAS proteins-each capable of transforming cells-are encoded when KRAS is activated by mutation2. No functional distinctions among the splice variants have so far been established. Oncogenic KRAS alters the metabolism of tumour cells3 in several ways, including increased glucose uptake and glycolysis even in the presence of abundant oxygen4 (the Warburg effect). Whereas these metabolic effects of oncogenic KRAS have been explained by transcriptional upregulation of glucose transporters and glycolytic enzymes3-5, it is not known whether there is direct regulation of metabolic enzymes. Here we report a direct, GTP-dependent interaction between KRAS4A and hexokinase 1 (HK1) that alters the activity of the kinase, and thereby establish that HK1 is an effector of KRAS4A. This interaction is unique to KRAS4A because the palmitoylation-depalmitoylation cycle of this RAS isoform enables colocalization with HK1 on the outer mitochondrial membrane. The expression of KRAS4A in cancer may drive unique metabolic vulnerabilities that can be exploited therapeutically.


Asunto(s)
Hexoquinasa/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regulación Alostérica , Animales , Línea Celular Tumoral , Activación Enzimática , Glucólisis , Guanosina Trifosfato/metabolismo , Hexoquinasa/química , Humanos , Técnicas In Vitro , Isoenzimas/metabolismo , Lipoilación , Masculino , Ratones , Mitocondrias/enzimología , Mitocondrias/metabolismo , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Neoplasias/enzimología , Neoplasias/metabolismo , Unión Proteica , Transporte de Proteínas
17.
J Med Chem ; 62(13): 6035-6046, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31181882

RESUMEN

Blockade of Ras activity by inhibiting its post-translational methylation catalyzed by isoprenylcysteine carboxylmethyltransferase (ICMT) has been suggested as a promising antitumor strategy. However, the paucity of inhibitors has precluded the clinical validation of this approach. In this work we report a potent ICMT inhibitor, compound 3 [UCM-1336, IC50 = 2 µM], which is selective against the other enzymes involved in the post-translational modifications of Ras. Compound 3 significantly impairs the membrane association of the four Ras isoforms, leading to a decrease of Ras activity and to inhibition of Ras downstream signaling pathways. In addition, it induces cell death in a variety of Ras-mutated tumor cell lines and increases survival in an in vivo model of acute myeloid leukemia. Because ICMT inhibition impairs the activity of the four Ras isoforms regardless of its activating mutation, compound 3 surmounts many of the common limitations of available Ras inhibitors described so far. In addition, these results validate ICMT as a valuable target for the treatment of Ras-driven tumors.


Asunto(s)
Alanina/uso terapéutico , Amidas/uso terapéutico , Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteína Metiltransferasas/antagonistas & inhibidores , Alanina/análogos & derivados , Alanina/síntesis química , Alanina/farmacología , Amidas/síntesis química , Amidas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Struct Mol Biol ; 26(7): 628-636, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209342

RESUMEN

Protein prenylation is believed to be catalyzed by three heterodimeric enzymes: FTase, GGTase1 and GGTase2. Here we report the identification of a previously unknown human prenyltransferase complex consisting of an orphan prenyltransferase α-subunit, PTAR1, and the catalytic ß-subunit of GGTase2, RabGGTB. This enzyme, which we named GGTase3, geranylgeranylates FBXL2 to allow its localization at cell membranes, where this ubiquitin ligase mediates the polyubiquitylation of membrane-anchored proteins. In cells, FBXL2 is specifically recognized by GGTase3 despite having a typical carboxy-terminal CaaX prenylation motif that is predicted to be recognized by GGTase1. Our crystal structure analysis of the full-length GGTase3-FBXL2-SKP1 complex reveals an extensive multivalent interface specifically formed between the leucine-rich repeat domain of FBXL2 and PTAR1, which unmasks the structural basis of the substrate-enzyme specificity. By uncovering a missing prenyltransferase and its unique mode of substrate recognition, our findings call for a revision of the 'prenylation code'.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Dimetilaliltranstransferasa/metabolismo , Proteínas F-Box/metabolismo , Transferasas Alquil y Aril/química , Línea Celular , Cristalografía por Rayos X , Dimetilaliltranstransferasa/química , Proteínas F-Box/química , Células HeLa , Humanos , Modelos Moleculares , Poliubiquitina/metabolismo , Conformación Proteica , Prenilación de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
19.
Proc Natl Acad Sci U S A ; 116(9): 3524-3529, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30733287

RESUMEN

RAP1-interacting adapter molecule (RIAM) mediates RAP1-induced integrin activation. The RAS-association (RA) segment of the RA-PH module of RIAM interacts with GTP-bound RAP1 and phosphoinositol 4,5 bisphosphate but this interaction is inhibited by the N-terminal segment of RIAM. Here we report the structural basis for the autoinhibition of RIAM by an intramolecular interaction between the IN region (aa 27-93) and the RA-PH module. We solved the crystal structure of IN-RA-PH to a resolution of 2.4-Å. The structure reveals that the IN segment associates with the RA segment and thereby suppresses RIAM:RAP1 association. This autoinhibitory configuration of RIAM can be released by phosphorylation at Tyr45 in the IN segment. Specific inhibitors of focal adhesion kinase (FAK) blocked phosphorylation of Tyr45, inhibited stimulated translocation of RIAM to the plasma membrane, and inhibited integrin-mediated cell adhesion in a Tyr45-dependent fashion. Our results reveal an unusual regulatory mechanism in small GTPase signaling by which the effector molecule is autoinhibited for GTPase interaction, and a modality of integrin activation at the level of RIAM through a FAK-mediated feedforward mechanism that involves reversal of autoinhibition by a tyrosine kinase associated with integrin signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Integrinas/química , Proteínas de la Membrana/química , Conformación Proteica , Proteínas de Unión a Telómeros/química , Proteínas Adaptadoras Transductoras de Señales/genética , Adhesión Celular/genética , Membrana Celular/química , Cristalografía por Rayos X , Proteína-Tirosina Quinasas de Adhesión Focal/química , Proteína-Tirosina Quinasas de Adhesión Focal/genética , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , Humanos , Proteínas de la Membrana/genética , Fosforilación , Complejo Shelterina , Transducción de Señal , Proteínas de Unión a Telómeros/genética
20.
Small GTPases ; 10(1): 20-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-28129035

RESUMEN

Ras is the best-studied member of the superfamily of small GTPases because of its role in cancer. Ras proteins transmit signals for proliferation, differentiation and survival. Three RAS genes encode 4 isoforms. All Ras isoforms have long been considered membrane bound, a localization required for function. Our recent study revealed that N-Ras differs from all other isoforms in being largely cytosolic even following modification with a prenyl lipid. Endogenous, cytosolic N-Ras chromatographed in both high and low molecular weight pools, a pattern that required prenylation, suggesting prenyl-dependent interaction with other proteins. VPS35, a coat protein of the retromer, was shown to interact with prenylated N-Ras in the cytosol. Silencing VPS35 results in partial N-Ras mislocalization on vesicular and tubulovesicular structures, reduced GTP-loading of Ras proteins, and inhibited proliferation and MAPK signaling in an oncogenic N-Ras-driven tumor cell line. Our data revealed a novel regulator of N-Ras trafficking and signaling.


Asunto(s)
Citosol/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas ras/metabolismo , Humanos , Prenilación de Proteína , Transporte de Proteínas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA