Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37645943

RESUMEN

Efforts to identify anti-cancer therapeutics and understand tumor-immune interactions are built with in vitro models that do not match the microenvironmental characteristics of human tissues. Using in vitro models which mimic the physical properties of healthy or cancerous tissues and a physiologically relevant culture medium, we demonstrate that the chemical and physical properties of the microenvironment regulate the composition and topology of the glycocalyx. Remarkably, we find that cancer and age-related changes in the physical properties of the microenvironment are sufficient to adjust immune surveillance via the topology of the glycocalyx, a previously unknown phenomenon observable only with a physiologically relevant culture medium.

2.
bioRxiv ; 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37333068

RESUMEN

Chemical screening studies have identified drug sensitivities across hundreds of cancer cell lines but most putative therapeutics fail to translate. Discovery and development of drug candidates in models that more accurately reflect nutrient availability in human biofluids may help in addressing this major challenge. Here we performed high-throughput screens in conventional versus Human Plasma-Like Medium (HPLM). Sets of conditional anticancer compounds span phases of clinical development and include non-oncology drugs. Among these, we characterize a unique dual-mechanism of action for brivudine, an agent otherwise approved for antiviral treatment. Using an integrative approach, we find that brivudine affects two independent targets in folate metabolism. We also traced conditional phenotypes for several drugs to the availability of nucleotide salvage pathway substrates and verified others for compounds that seemingly elicit off-target anticancer effects. Our findings establish generalizable strategies for exploiting conditional lethality in HPLM to reveal therapeutic candidates and mechanisms of action.

3.
Mol Ther Methods Clin Dev ; 24: 380-393, 2022 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-35284590

RESUMEN

Ex vivo expansion conditions used to generate T cells for immunotherapy are thought to adopt metabolic phenotypes that impede therapeutic efficacy in vivo. The comparison of five different culture media used for clinical T cell expansion revealed unique optima based on different output variables, including proliferation, differentiation, function, activation, and mitochondrial phenotypes. The extent of proliferation and function depended on the culture media rather than stimulation conditions. Moreover, the expanded T cell end products adapted their metabolism when switched to a different media formulation, as shown by glucose and glutamine uptake and patterns of glucose isotope labeling. However, adoption of these metabolic phenotypes was uncoupled to T cell function. Expanded T cell products cultured in ascites from ovarian cancer patients displayed suppressed mitochondrial activity and function irrespective of the ex vivo expansion media. Thus, ex vivo T cell expansion media have profound impacts on metabolism and function.

5.
Methods Mol Biol ; 2377: 29-42, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34709609

RESUMEN

Forward genetic screens across hundreds of cancer cell lines have started to define the genetic dependencies of proliferating human cells. However, most such screens have been performed in vitro with little consideration into how medium composition might affect gene essentiality. This protocol describes a method to use CRISPR/Cas9-based loss-of-function screens to ask how gene essentiality in human cell lines varies with medium composition. First, a single-guide RNA (sgRNA) library is packaged into lentivirus, and an optimal infection titer is determined for the target cells. Following selection, genomic DNA (gDNA) is extracted from an aliquot of the transduced cells. The remaining transduced cells are then screened in at least two distinct cell culture media. At the conclusion of the screening period, gDNA is collected from each cell population. Next, high-throughput sequencing is used to determine sgRNA barcode abundances from the initial and each of the final populations. Finally, an analytical pipeline is used to identify medium-essential candidate genes from these screen results.


Asunto(s)
Sistemas CRISPR-Cas , Genes Esenciales , Sistemas CRISPR-Cas/genética , Línea Celular , ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , ARN Guía de Kinetoplastida/genética
6.
Cell Metab ; 33(6): 1248-1263.e9, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33651980

RESUMEN

Forward genetic screens across hundreds of cancer cell lines have started to define the genetic dependencies of proliferating human cells and how these vary by genotype and lineage. Most screens, however, have been carried out in culture media that poorly reflect metabolite availability in human blood. Here, we performed CRISPR-based screens in traditional versus human plasma-like medium (HPLM). Sets of conditionally essential genes in human cancer cell lines span several cellular processes and vary with both natural cell-intrinsic diversity and the combination of basal and serum components that comprise typical media. Notably, we traced the causes for each of three conditional CRISPR phenotypes to the availability of metabolites uniquely defined in HPLM versus conventional media. Our findings reveal the profound impact of medium composition on gene essentiality in human cells, and also suggest general strategies for using genetic screens in HPLM to uncover new cancer vulnerabilities and gene-nutrient interactions.


Asunto(s)
Sistemas CRISPR-Cas , Medios de Cultivo , Línea Celular Tumoral , Humanos
7.
Cancer Cell ; 39(1): 28-37, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33125860

RESUMEN

Immune cells' metabolism influences their differentiation and function. Given that a complex interplay of environmental factors within the tumor microenvironment (TME) can have a profound impact on the metabolic activities of immune, stromal, and tumor cell types, there is emerging interest to advance understanding of these diverse metabolic phenotypes in the TME. Here, we discuss cell-extrinsic contributions to the metabolic activities of immune cells. Then, considering recent technical advances in experimental systems and metabolic profiling technologies, we propose future directions to better understand how immune cells meet their metabolic demands in the TME, which can be leveraged for therapeutic benefit.


Asunto(s)
Sistema Inmunológico/metabolismo , Neoplasias/inmunología , Animales , Humanos , Metabolómica , Microambiente Tumoral
8.
iScience ; 23(1): 100759, 2020 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-31887663

RESUMEN

T lymphocytes are critical for effective immunity, and the ability to study their behavior in vitro can facilitate major insights into their development, function, and fate. However, the composition of human plasma differs from conventional media, and we hypothesized that such differences could impact immune cell physiology. Here, we showed that relative to the medium typically used to culture lymphocytes (RPMI), a physiologic medium (human plasma-like medium; HPLM) induced markedly different transcriptional responses in human primary T cells and in addition, improved their activation upon antigen stimulation. We found that this medium-dependent effect on T cell activation is linked to Ca2+, which is six-fold higher in HPLM than in RPMI. Thus, a medium that more closely resembles human plasma has striking effects on T cell biology, further demonstrates that medium composition can profoundly affect experimental results, and broadly suggests that physiologic media may offer a valuable way to study cultured immune cells.

9.
Trends Cell Biol ; 29(11): 854-861, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31623927

RESUMEN

Developed decades ago, traditional culture media were not intended to resemble the metabolic composition of human blood, and indeed poorly do so. Yet, despite what is now a clear recognition that environmental factors influence metabolism, such media remain standard to in vitro studies across virtually all areas of biological research. The recent development of physiologic media, like other efforts designed to address the modeling capacity of cell culture, holds immense potential to improve understanding and interpretation of diverse biological and pharmacological studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Animales , Línea Celular , Humanos , Ratones , Plasma/química
10.
Nature ; 559(7715): 632-636, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29995852

RESUMEN

The chemotherapeutic drug methotrexate inhibits the enzyme dihydrofolate reductase1, which generates tetrahydrofolate, an essential cofactor in nucleotide synthesis2. Depletion of tetrahydrofolate causes cell death by suppressing DNA and RNA production3. Although methotrexate is widely used as an anticancer agent and is the subject of over a thousand ongoing clinical trials4, its high toxicity often leads to the premature termination of its use, which reduces its potential efficacy5. To identify genes that modulate the response of cancer cells to methotrexate, we performed a CRISPR-Cas9-based screen6,7. This screen yielded FTCD, which encodes an enzyme-formimidoyltransferase cyclodeaminase-that is required for the catabolism of the amino acid histidine8, a process that has not previously been linked to methotrexate sensitivity. In cultured cancer cells, depletion of several genes in the histidine degradation pathway markedly decreased sensitivity to methotrexate. Mechanistically, histidine catabolism drains the cellular pool of tetrahydrofolate, which is particularly detrimental to methotrexate-treated cells. Moreover, expression of the rate-limiting enzyme in histidine catabolism is associated with methotrexate sensitivity in cancer cell lines and with survival rate in patients. In vivo dietary supplementation of histidine increased flux through the histidine degradation pathway and enhanced the sensitivity of leukaemia xenografts to methotrexate. The histidine degradation pathway markedly influences the sensitivity of cancer cells to methotrexate and may be exploited to improve methotrexate efficacy through a simple dietary intervention.


Asunto(s)
Histidina/metabolismo , Metotrexato/farmacología , Metotrexato/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Amoníaco-Liasas/deficiencia , Amoníaco-Liasas/genética , Amoníaco-Liasas/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Femenino , Antagonistas del Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/uso terapéutico , Glutamato Formimidoiltransferasa/deficiencia , Glutamato Formimidoiltransferasa/genética , Glutamato Formimidoiltransferasa/metabolismo , Histidina/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Enzimas Multifuncionales , Nucleótidos/biosíntesis , Proteína Portadora de Folato Reducido/genética , Proteína Portadora de Folato Reducido/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Tetrahidrofolatos/deficiencia , Tetrahidrofolatos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cell ; 169(2): 258-272.e17, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388410

RESUMEN

A complex interplay of environmental factors impacts the metabolism of human cells, but neither traditional culture media nor mouse plasma mimic the metabolite composition of human plasma. Here, we developed a culture medium with polar metabolite concentrations comparable to those of human plasma (human plasma-like medium [HPLM]). Culture in HPLM, relative to that in traditional media, had widespread effects on cellular metabolism, including on the metabolome, redox state, and glucose utilization. Among the most prominent was an inhibition of de novo pyrimidine synthesis-an effect traced to uric acid, which is 10-fold higher in the blood of humans than of mice and other non-primates. We find that uric acid directly inhibits uridine monophosphate synthase (UMPS) and consequently reduces the sensitivity of cancer cells to the chemotherapeutic agent 5-fluorouracil. Thus, media that better recapitulates the composition of human plasma reveals unforeseen metabolic wiring and regulation, suggesting that HPLM should be of broad utility.


Asunto(s)
Medios de Cultivo/química , Complejos Multienzimáticos/antagonistas & inhibidores , Orotato Fosforribosiltransferasa/antagonistas & inhibidores , Orotidina-5'-Fosfato Descarboxilasa/antagonistas & inhibidores , Ácido Úrico/metabolismo , Anciano , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Fluorouracilo/farmacología , Glucosa/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Persona de Mediana Edad , Complejos Multienzimáticos/química , Orotato Fosforribosiltransferasa/química , Orotidina-5'-Fosfato Descarboxilasa/química , Dominios Proteicos , Pirimidinas/biosíntesis
12.
Science ; 351(6268): 43-8, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26449471

RESUMEN

Leucine is a proteogenic amino acid that also regulates many aspects of mammalian physiology, in large part by activating the mTOR complex 1 (mTORC1) protein kinase, a master growth controller. Amino acids signal to mTORC1 through the Rag guanosine triphosphatases (GTPases). Several factors regulate the Rags, including GATOR1, aGTPase-activating protein; GATOR2, a positive regulator of unknown function; and Sestrin2, a GATOR2-interacting protein that inhibits mTORC1 signaling. We find that leucine, but not arginine, disrupts the Sestrin2-GATOR2 interaction by binding to Sestrin2 with a dissociation constant of 20 micromolar, which is the leucine concentration that half-maximally activates mTORC1. The leucine-binding capacity of Sestrin2 is required for leucine to activate mTORC1 in cells. These results indicate that Sestrin2 is a leucine sensor for the mTORC1 pathway.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Leucina/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Redes y Vías Metabólicas , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Proteínas/química , Transducción de Señal
13.
Nature ; 520(7547): 363-7, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25855294

RESUMEN

Cancer cells adapt their metabolic processes to support rapid proliferation, but less is known about how cancer cells alter metabolism to promote cell survival in a poorly vascularized tumour microenvironment. Here we identify a key role for serine and glycine metabolism in the survival of brain cancer cells within the ischaemic zones of gliomas. In human glioblastoma multiforme, mitochondrial serine hydroxymethyltransferase (SHMT2) and glycine decarboxylase (GLDC) are highly expressed in the pseudopalisading cells that surround necrotic foci. We find that SHMT2 activity limits that of pyruvate kinase (PKM2) and reduces oxygen consumption, eliciting a metabolic state that confers a profound survival advantage to cells in poorly vascularized tumour regions. GLDC inhibition impairs cells with high SHMT2 levels as the excess glycine not metabolized by GLDC can be converted to the toxic molecules aminoacetone and methylglyoxal. Thus, SHMT2 is required for cancer cells to adapt to the tumour environment, but also renders these cells sensitive to glycine cleavage system inhibition.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Glicina Hidroximetiltransferasa/metabolismo , Glicina/metabolismo , Isquemia/metabolismo , Acetona/análogos & derivados , Acetona/metabolismo , Acetona/toxicidad , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/enzimología , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Femenino , Glioblastoma/irrigación sanguínea , Glioblastoma/enzimología , Glicina-Deshidrogenasa (Descarboxilante)/antagonistas & inhibidores , Glicina-Deshidrogenasa (Descarboxilante)/metabolismo , Humanos , Isquemia/enzimología , Isquemia/patología , Ratones , Necrosis , Consumo de Oxígeno , Piruvaldehído/metabolismo , Piruvaldehído/toxicidad , Piruvato Quinasa/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171410

RESUMEN

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Asunto(s)
Transición Epitelial-Mesenquimal , Pirimidinas/metabolismo , Animales , Carcinoma/metabolismo , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , ARN Interferente Pequeño/metabolismo
15.
Cancer Discov ; 2(10): 881-98, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23009760

RESUMEN

Cancer cells must rewire cellular metabolism to satisfy the demands of growth and proliferation. Although many of the metabolic alterations are largely similar to those in normal proliferating cells, they are aberrantly driven in cancer by a combination of genetic lesions and nongenetic factors such as the tumor microenvironment. However, a single model of altered tumor metabolism does not describe the sum of metabolic changes that can support cell growth. Instead, the diversity of such changes within the metabolic program of a cancer cell can dictate by what means proliferative rewiring is driven, and can also impart heterogeneity in the metabolic dependencies of the cell. A better understanding of this heterogeneity may enable the development and optimization of therapeutic strategies that target tumor metabolism.


Asunto(s)
Metabolismo Energético/fisiología , Neoplasias/metabolismo , Proliferación Celular , Metabolismo Energético/genética , Glucólisis/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/genética , Transcripción Genética , Microambiente Tumoral
16.
ACS Chem Biol ; 7(11): 1840-7, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22891768

RESUMEN

The human asparaginase-like protein 1 (hASRGL1) catalyzes the hydrolysis of l-asparagine and isoaspartyl-dipeptides. As an N-terminal nucleophile (Ntn) hydrolase superfamily member, the active form of hASRGL1 is generated by an intramolecular cleavage step with Thr168 as the catalytic residue. However, in vitro, autoprocessing is incomplete (~50%), fettering the biophysical characterization of hASRGL1. We circumvented this obstacle by constructing a circularly permuted hASRGL1 that uncoupled the autoprocessing reaction, allowing us to kinetically and structurally characterize this enzyme and the precursor-like hASRGL1-Thr168Ala variant. Crystallographic and biochemical evidence suggest an activation mechanism where a torsional restraint on the Thr168 side chain helps drive the intramolecular processing reaction. Cleavage and formation of the active site releases the torsional restriction on Thr168, which is facilitated by a small conserved Gly-rich loop near the active site that allows the conformational changes necessary for activation.


Asunto(s)
Amidohidrolasas/química , Amidohidrolasas/metabolismo , Asparaginasa/química , Asparaginasa/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Amidohidrolasas/genética , Asparaginasa/genética , Asparagina/metabolismo , Autoantígenos/genética , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática , Humanos , Hidrólisis , Modelos Moleculares , Mutación Puntual , Conformación Proteica
17.
Methods Enzymol ; 502: 291-319, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22208990

RESUMEN

Cancer has become the leading cause of death in the developed world and has remained one of the most difficult diseases to treat. One of the difficulties in treating cancer is that conventional chemotherapies often have unacceptable toxicities toward normal cells at the doses required to kill tumor cells. Thus, the demand for new and improved tumor specific therapeutics for the treatment of cancer remains high. Alterations to cellular metabolism constitute a nearly universal feature of many types of cancer cells. In particular, many tumors exhibit deficiencies in one or more amino acid synthesis or salvage pathways forcing a reliance on the extracellular pool of these amino acids to satisfy protein biosynthesis demands. Therefore, one treatment modality that satisfies the objective of developing cancer cell-selective therapeutics is the systemic depletion of that tumor-essential amino acid, which can result in tumor apoptosis with minimal side effects to normal cells. While this strategy was initially suggested over 50 years ago, it has been recently experiencing a renaissance owing to advances in protein engineering technology, and more sophisticated approaches to studying the metabolic differences between tumorigenic and normal cells. Dietary restriction is typically not sufficient to achieve a therapeutically relevant level of amino acid depletion for cancer treatment. Therefore, intravenous administration of enzymes is used to mediate the degradation of such amino acids for therapeutic purposes. Unfortunately, the human genome does not encode enzymes with the requisite catalytic or pharmacological properties necessary for therapeutic purposes. The use of heterologous enzymes has been explored extensively both in animal studies and in clinical trials. However, heterologous enzymes are immunogenic and elicit adverse responses ranging from anaphylactic shock to antibody-mediated enzyme inactivation, and therefore have had limited utility. The one notable exception is Escherichia colil-asparaginase II (EcAII), which has been FDA-approved for the treatment of childhood acute lymphoblastic leukemia. The use of engineered human enzymes, to which natural tolerance is likely to prevent recognition by the adaptive immune system, offers a novel approach for capitalizing on the promising strategy of systemic depletion of tumor-essential amino acids. In this work, we review several strategies that we have developed to: (i) reduce the immunogenicity of a nonhuman enzyme, (ii) engineer human enzymes for novel catalytic specificities, and (iii) improve the pharmacological characteristics of a human enzyme that exhibits the requisite substrate specificity for amino acid degradation but exhibits low activity and stability under physiological conditions.


Asunto(s)
Aminoácidos/deficiencia , Asparaginasa/administración & dosificación , Terapia Enzimática/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Animales , Asparaginasa/genética , Asparaginasa/inmunología , Asparaginasa/uso terapéutico , Sitios de Unión , Clonación Molecular , Simulación por Computador , Estabilidad de Enzimas , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Cinética , Ratones , Mutagénesis Sitio-Dirigida , Polietilenglicoles/química , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
18.
Biochemistry ; 50(14): 3025-33, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21375249

RESUMEN

The enzymatic deamidation of N-terminal L-Asn by N-terminal asparagine amidohydrolase (NTAN1) is a feature of the ubiquitin-dependent N-end rule pathway of protein degradation, which relates the in vivo half-life of a protein to the identity of its N-terminal residue. Herein, we report the bacterial expression, purification, and biochemical characterization of human NTAN1 (hNTAN1). We show here that hNTAN1 is highly selective for the hydrolysis of N-terminal peptidyl L-Asn but fails to deamidate free L-Asn or L-Gln, N-terminal peptidyl L-Gln, or acetylated N-terminal peptidyl L-Asn. Similar to other N-terminal deamidases, hNTAN1 is shown to possess a critical Cys residue that is absolutely required for catalysis, corroborated in part by abolishment of activity through the Cys75Ala point mutation. We also present evidence that the exposure of a conserved L-Pro at the N-terminus of hNTAN1 following removal of the initiating L-Met is important for the function of the enzyme. The results presented here should assist in the elucidation of molecular mechanisms underlying the neurological defects of NTAN1-deficient mice observed in other studies, and in the discovery of potential physiological substrates targeted by the enzyme in the modulation of protein turnover via the N-end rule pathway.


Asunto(s)
Amidohidrolasas/metabolismo , Asparagina/metabolismo , Ácido Aspártico/metabolismo , Proteínas Recombinantes/metabolismo , Amidohidrolasas/química , Amidohidrolasas/genética , Animales , Asparagina/química , Ácido Aspártico/química , Biocatálisis/efectos de los fármacos , Dicroismo Circular , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Electroforesis en Gel de Poliacrilamida , Humanos , Concentración de Iones de Hidrógeno , Cinética , Metales/farmacología , Ratones , Modelos Químicos , Estructura Molecular , Mutación , Proteínas Recombinantes/química , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato
19.
Proc Natl Acad Sci U S A ; 108(4): 1272-7, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21209329

RESUMEN

A number of heterologous enzymes have been investigated for cancer treatment and other therapeutic applications; however, immunogenicity issues have limited their clinical utility. Here, a new approach has been created for heterologous enzyme deimmunization whereby combinatorial saturation mutagenesis is coupled with a screening strategy that capitalizes on the evolutionary biology concept of neutral drift, and combined with iterative computational prediction of T-cell epitopes to achieve extensive reengineering of a protein sequence for reduced MHC-II binding propensity without affecting catalytic and pharmacological properties. Escherichia coli L-asparaginase II (EcAII), the only nonhuman enzyme approved for repeated administration, is critical in treatment of childhood acute lymphoblastic leukemia (ALL), but elicits adverse antibody responses in a significant fraction of patients. The neutral drift screening of combinatorial saturation mutagenesis libraries at a total of 12 positions was used to isolate an EcAII variant containing eight amino acid substitutions within computationally predicted T-cell epitopes--of which four were nonconservative--while still exhibiting k(cat)/K(M) = 10(6) M(-1) s(-1) for L-Asn hydrolysis. Further, immunization of HLA-transgenic mice expressing the ALL-associated DRB1*0401 allele with the engineered variant resulted in significantly reduced T-cell responses and a 10-fold reduction in anti-EcAII IgG titers relative to the existing therapeutic. This significant reduction in the immunogenicity of EcAII may be clinically relevant for ALL treatment and illustrates the potential of employing neutral drift screens to achieve large jumps in sequence space as may be required for the deimmunization of heterologous proteins.


Asunto(s)
Asparaginasa/inmunología , Epítopos de Linfocito T/inmunología , Proteínas de Escherichia coli/inmunología , Inmunización/métodos , Animales , Asparaginasa/química , Asparaginasa/genética , Dominio Catalítico , Biología Computacional/métodos , Evolución Molecular Dirigida , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Femenino , Citometría de Flujo , Flujo Genético , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Cadenas HLA-DRB1 , Humanos , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Transgénicos , Modelos Moleculares , Mutación , Biblioteca de Péptidos , Estructura Terciaria de Proteína , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
Biochemistry ; 48(46): 11026-31, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19839645

RESUMEN

Herein we report the bacterial expression, purification, and enzymatic characterization of the human asparaginase-like protein 1 (hASRGL1). We present evidence that hASRGL1 exhibits beta-aspartyl peptidase activity consistent with enzymes designated as plant-type asparaginases, which had thus far been found in only plants and bacteria. Similar to nonmammalian plant-type asparaginases, hASRGL1 is shown to be an Ntn hydrolase for which Thr168 serves as the essential N-terminal nucleophile for intramolecular processing and catalysis, corroborated in part by abolishment of both activities through the Thr168Ala point mutation. In light of the activity profile reported here, ASRGL1s may act synergistically with protein l-isoaspartyl methyl transferase to relieve accumulation of potentially toxic isoaspartyl peptides in mammalian brain and other tissues.


Asunto(s)
Amidohidrolasas/metabolismo , Asparaginasa/metabolismo , Autoantígenos/metabolismo , Dipeptidasas/química , Amidohidrolasas/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Asparaginasa/química , Asparaginasa/genética , Autoantígenos/química , Autoantígenos/genética , Biocatálisis , Dominio Catalítico , Dipéptidos/química , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA