Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(6): e0285971, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37267316

RESUMO

Antibody-based therapeutics and recombinant protein reagents are often produced in mammalian expression systems, which provide human-like post-translational modifications. Among the available mammalian cell lines used for recombinant protein expression, Chinese hamster ovary (CHO)-derived suspension cells are generally utilized because they are easy to culture and tend to produce proteins in high yield. However, some proteins purified from CHO cell overexpression suffer from clipping and display undesired non-human post translational modifications (PTMs). In addition, CHO cell lines are often not suitable for producing proteins with many glycosylation motifs for structural biology studies, as N-linked glycosylation of proteins poses challenges for structure determination by X-ray crystallography. Hence, alternative and complementary cell lines are required to address these issues. Here, we present a robust method for expressing proteins in human embryonic kidney 293 (HEK293)-derived stable pools, leading to recombinant protein products with much less clipped species compared to those expressed in CHO cells and with higher yield compared to those expressed in transiently-transfected HEK293 cells. Importantly, the stable pool generation protocol is also applicable to HEK293S GnTI- (N-acetylglucosaminyltransferase I-negative) and Expi293F GnTI- suspension cells, facilitating production of high yields of proteins with less complex glycans for use in structural biology projects. Compared to HEK293S GnTI- stable pools, Expi293F GnTI- stable pools consistently produce proteins with similar or higher expression levels. HEK293-derived stable pools can lead to a significant cost reduction and greatly promote the production of high-quality proteins for diverse research projects.


Assuntos
Rim , Cricetinae , Animais , Humanos , Cricetulus , Células HEK293 , Células CHO , Proteínas Recombinantes/química , Rim/metabolismo
2.
Sci Transl Med ; 13(608)2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433637

RESUMO

Therapeutic approaches are needed to promote T cell-mediated destruction of poorly immunogenic, "cold" tumors typically associated with minimal response to immune checkpoint blockade (ICB) therapy. Bispecific T cell engager (BiTE) molecules induce redirected lysis of cancer cells by polyclonal T cells and have demonstrated promising clinical activity against solid tumors in some patients. However, little is understood about the key factors that govern clinical responses to these therapies. Using an immunocompetent mouse model expressing a humanized CD3ε chain (huCD3e mice) and BiTE molecules directed against mouse CD19, mouse CLDN18.2, or human EPCAM antigens, we investigated the pharmacokinetic and pharmacodynamic parameters and immune correlates associated with BiTE efficacy across multiple syngeneic solid-tumor models. These studies demonstrated that pretreatment tumor-associated T cell density is a critical determinant of response to BiTE therapy, identified CD8+ T cells as important targets and mediators of BiTE activity, and revealed an antagonistic role for CD4+ T cells in BiTE efficacy. We also identified therapeutic combinations, including ICB and 4-1BB agonism, that synergized with BiTE treatment in poorly T cell-infiltrated, immunotherapy-refractory tumors. In these models, BiTE efficacy was dependent on local expansion of tumor-associated CD8+ T cells, rather than their recruitment from circulation. Our findings highlight the relative contributions of baseline T cell infiltration, local T cell proliferation, and peripheral T cell trafficking for BiTE molecule-mediated efficacy, identify combination strategies capable of overcoming resistance to BiTE therapy, and have clinical relevance for the development of BiTE and other T cell engager therapies.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Animais , Anticorpos Biespecíficos/uso terapêutico , Antígenos CD19 , Complexo CD3 , Linfócitos T CD8-Positivos , Claudinas , Humanos , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico
3.
Artigo em Inglês | MEDLINE | ID: mdl-31841981

RESUMO

There is a great need for high-throughput protein purification to produce protein molecules for research and therapeutics. Although there have been significant advancements made in automated multi-step chromatography and preparative in-process design-of-experiment (DOE) capabilities in commercial fast performance liquid chromatography (FPLC) instruments, almost all commercial FPLCs rely on a binary buffer mixing system, which hinders automated buffer preparation. Nevertheless, current-generation FPLCs are equipped with a quaternary mixer designed for limited in-line buffer preparation and preparative pH scouting DOE experiments. We decided to leverage the quaternary mixing capability by extending and re-programming AkTA Avant's quaternary valve into an automated in-process buffer preparation system to simplify automated purification requiring complex washing steps. We accomplished this by using two extra inlet valves, a sample valve, and versatile valve to split inputs of the quaternary valve into software-selectable stock solutions of pH buffers, salts, eluents, and additives. We also devised a new flow scheme to perform automated two-step chromatography using only one versatile valve. This was accomplished by using only stock parts and software to facilitate reproduction. To demonstrate the versatility and capability of the system, we purified a transmembrane protein that requires a detergent to stay soluble and needs an in-column, high-salt washing step to achieve high purity.


Assuntos
Automação Laboratorial/instrumentação , Membrana Celular/química , Cromatografia Líquida/instrumentação , Proteínas de Membrana/isolamento & purificação , Soluções Tampão , Cromatografia Líquida/métodos , Desenho de Equipamento , Humanos
4.
Exp Cell Res ; 370(2): 208-226, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29940176

RESUMO

STEAP2 is a member of the Six-Transmembrane Epithelial Antigen of the Prostate (STEAP) protein family that is proposed to function as metalloreductase. While STEAP2 shows a complex subcellular distribution pattern localizing to both secretory and endocytic pathway organelles, how such broad steady-state distribution is maintained is unknown. Similarly, whether STEAP2 undergoes any compartment-specific modulation during intracellular trafficking has not been reported. Leveraging a newly-identified monoclonal antibody that recognizes a conformation-sensitive epitope nested in the second extracellular loop of STEAP2, we demonstrate that the epitope formation was dependent on the cholesterol content of the membrane in which STEAP2 was embedded. Monitoring the STEAP2-dependent internalization of this antibody uncovered STEAP2's rapid internalization from the cell surface and their subsequence trafficking to the Golgi region and endosome-like puncta. Acute inhibition of endocytosis also increased the detectable amount of STEAP2 at the plasma membrane. Collectively, these experiments demonstrate that an intricate balance of membrane flux between the secretory and endocytic pathways underlies the characteristic broad subcellular localization of STEAP2. By using a cell-based assay that detects the metalloreductase functions of cell surface-localizing STEAP4, STEAP2's metalloreductase activities were not detectable, suggesting that its enzymatic function is suppressed at the plasma membrane. The conformational modulation of STEAP2 by the local membrane cholesterol content can therefore serve as a potential mechanism to modulate STEAP2 function in a compartment-restricted manner, by coupling a pre-existing difference in cholesterol content among different cellular membranes to a dynamic trafficking process leading to broad subcellular distribution.


Assuntos
Antígenos de Neoplasias/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Oxirredutases/metabolismo , Animais , Transporte Biológico/fisiologia , Movimento Celular/fisiologia , Endocitose/fisiologia , Endossomos/metabolismo , Humanos , Camundongos , Conformação Molecular , Transporte Proteico
5.
J Orthop Res ; 36(4): 1238-1247, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28971529

RESUMO

Designing drugs to treat diseases associated with articular joints, particularly those targeting chondrocytes, is challenging due to unique local environmental constraints including the avascular nature of cartilage, the absence of a closed joint compartment, and a highly cross-linked extracellular matrix. In an effort to address these challenges, we developed a novel strategy to prolong residence time of intra-articularly administered protein therapeutics. Avimer domains are naturally found in membrane polypeptides and mediate diverse protein-protein interactions. Screening of a phage Avimer domain library led to identification of several low affinity type II collagen-binding Avimers. Following several rounds of mutagenesis and reselection, these initial hits were transformed to high affinity, selective type II collagen-binding Avimers. One such Avimer (M26) persisted in rat knees for at least 1 month following intra-articular administration. Fusion of this Avimer to a candidate therapeutic payload, IL-1Ra, yielded a protein construct which simultaneously bound to type II collagen and to IL-1 receptor. In vitro, IL-1Ra_M26 bound selectively to cartilage explants and remained associated even after extensive washing. Binding appeared to occur preferentially to pericellular regions surrounding chondrocytes. An acute intra-articular IL-1-induced IL-6 challenge rat model was employed to assess in vivo pharmacodynamics. Whereas both IL-1Ra_M26 and native IL-1Ra inhibited IL-6 output when co-administered with the IL-1 challenge, only IL-1Ra_M26 inhibited when administered 1 week prior to IL-1 challenge. Collagen-binding Avimers thus represent a promising strategy for enhancing cartilage residence time of protein therapeutics. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1238-1247, 2018.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Artropatias/tratamento farmacológico , Proteínas/administração & dosagem , Animais , Colágeno Tipo II/metabolismo , Feminino , Humanos , Injeções Intra-Articulares , Masculino , Domínios Proteicos , Engenharia de Proteínas , Ratos Endogâmicos Lew , Ratos Sprague-Dawley
6.
Protein Expr Purif ; 137: 7-12, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28619526

RESUMO

Pichia pastoris is a highly successful recombinant protein expression system due to its ability to quickly generate large quantities of recombinant proteins in simple media. P. pastoris has been used to successfully generate milligram quantities of many important human membrane proteins, including G-protein coupled receptors, ion channels, and transporters, which are becoming increasingly important therapeutic targets. Despite these successes, protein expression in P. pastoris is still cumbersome due to a need to change growth media from glycerol media to methanol induction media, which minimizes inhibition of the AOX1 promoter by residual glycerol. Taking advantage of this behavior of the AOX1 promoter, we developed Buffered extra-YNB Glycerol Methanol (BYGM) auto-induction media (100 mM potassium phosphate pH 6.0, 2.68% w/v YNB, 0.4% v/v glycerol, 0.5% v/v methanol, and 8 × 10-5% w/v biotin) which not only simplified the protein expression process, but also optimized protein expression levels in P. pastoris. We successfully used this auto-induction method to overexpress the target in both MutS and Mut+ strains. Moreover, we show that this method can facilitate screening high-expressing clones, as well as enable parallel protein production in P. pastoris.


Assuntos
Membrana Celular , Proteínas Fúngicas , Expressão Gênica , Proteínas de Membrana , Pichia , Regiões Promotoras Genéticas , Membrana Celular/genética , Membrana Celular/metabolismo , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Pichia/genética , Pichia/metabolismo
7.
PLoS One ; 8(4): e61432, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23630589

RESUMO

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and ß-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/ß-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and ß-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


Assuntos
Fármacos Antiobesidade/farmacologia , Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas de Membrana/antagonistas & inibidores , Obesidade/tratamento farmacológico , Peptídeos/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Fármacos Antiobesidade/farmacocinética , Sítios de Ligação , Ligação Competitiva , Peso Corporal/efeitos dos fármacos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Fatores de Crescimento de Fibroblastos/química , Insulina/sangue , Proteínas Klotho , Macaca fascicularis , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Mimetismo Molecular , Dados de Sequência Molecular , Obesidade/sangue , Peptídeos/farmacocinética , Ligação Proteica , Ratos , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/química , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/biossíntese , Albumina Sérica/farmacocinética , Albumina Sérica/farmacologia , Transdução de Sinais , Triglicerídeos/sangue
8.
Proc Natl Acad Sci U S A ; 108(5): 2106-11, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21245315

RESUMO

In Escherichia coli, the σ(E) transcription factor monitors and maintains outer membrane (OM) integrity by activating genes required for assembly of its two key components, outer membrane proteins (OMPs) and lipopolysaccharide (LPS) and by transcribing small RNAs to down-regulate excess unassembled OMPs. σ(E) activity is governed by the rate of degradation of its membrane-spanning anti-σ factor, RseA. Importantly, the DegS protease can initiate RseA cleavage only when activated by binding to unassembled OMPs. The prevalent paradigm has been that the σ(E) response is controlled by the amount of activated DegS. Here we demonstrate that inactivation of a second negative regulator, the periplasmic protein RseB, is also required for σ(E) induction in vivo. Moreover, OMPs, previously known only to activate DegS, also generate a signal to antagonize RseB inhibition. This signal may be lipid related, as RseB is structurally similar to proteins that bind lipids. We propose that the use of an AND gate enables σ(E) to sense and integrate multivariate signals from the envelope.


Assuntos
Proteínas de Escherichia coli/fisiologia , Escherichia coli/fisiologia , Proteínas de Membrana/fisiologia , Transdução de Sinais , Proteínas da Membrana Bacteriana Externa/fisiologia , Membrana Celular/fisiologia
9.
Genes Dev ; 18(21): 2686-97, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15520285

RESUMO

Proteolytic cascades are widely implicated in signaling between cellular compartments. In Escherichia coli, accumulation of unassembled outer membrane porins (OMPs) in the envelope leads to expression of sigma(E)-dependent genes in the cytoplasmic cellular compartment. A proteolytic cascade conveys the OMP signal by regulated proteolysis of RseA, a membrane-spanning anti-sigma factor whose cytoplasmic domain inhibits sigma(E)-dependent transcription. Upon activation by OMP C termini, the membrane localized DegS protease cleaves RseA in its periplasmic domain, the membrane-embedded protease RseP (YaeL) cleaves RseA near the inner membrane, and the released cytoplasmic RseA fragment is further degraded. Initiation of RseA degradation by activated DegS makes the system sensitive to a wide range of OMP concentrations and unresponsive to variations in the levels of DegS and RseP proteases. These features rely on the inability of RseP to cleave intact RseA. In the present report, we demonstrate that RseB, which binds to the periplasmic face of RseA, and DegS each independently inhibits RseP cleavage of intact RseA. Thus, the function of RseB, widely conserved among bacteria using the sigma(E) pathway, and the second role of DegS (in addition to RseA proteolysis initiation) is to improve the performance characteristics of this signal transduction system.


Assuntos
Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Fator sigma/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Sequência de Bases , Endopeptidases/genética , Endopeptidases/metabolismo , Escherichia coli/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Membrana/genética , Dados de Sequência Molecular , Porinas/genética , Porinas/metabolismo , Fator sigma/genética , Fatores de Transcrição/genética
10.
Mol Microbiol ; 52(3): 613-9, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15101969

RESUMO

The Escherichia colisigma(E)-dependent stress response pathway controls the expression of genes encoding periplasmic folding catalysts, proteases, biosynthesis enzymes for lipid A (a component of lipopolysaccharide or LPS) and other proteins known or predicted to function in or produce components of the envelope. When E. coli is subjected to heat or other stresses that generate unfolded envelope proteins, sigma(E) activity is induced. Four key players in this signal transduction pathway have been identified: RseA, an inner membrane sigma(E) antisigma factor; RseB, a periplasmic protein that binds to the periplasmic face of RseA; and the DegS and YaeL proteases. The major point of regulation, the interaction between sigma(E) and RseA, is primarily controlled by the stability of RseA. Envelope stress promotes RseA degradation, which occurs by a proteolytic cascade initiated by DegS. There is evidence that one sigma(E)-inducing stress (OmpC overexpression) directly activates DegS to cleave RseA. Secondarily, envelope stress may relieve RseB-mediated enhancement of RseA activity. Additional levels of control upon sigma(E) activity may become evident upon further study of this stress response pathway.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Resposta ao Choque Térmico , Fator sigma/metabolismo , Fatores de Transcrição/metabolismo , Animais , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana/metabolismo , Dobramento de Proteína , Transdução de Sinais
11.
Cell ; 113(1): 61-71, 2003 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-12679035

RESUMO

Transmembrane signaling between intracellular compartments is often controlled by regulated proteolysis. Escherichia coli respond to misfolded or unfolded outer-membrane porins (OMPs) in the periplasm by inducing sigma(E)-dependent transcription of stress genes in the cytoplasm. This process requires a proteolytic cascade initiated by the DegS protease, which destroys a transmembrane protein (RseA) that normally binds to and inhibits sigma(E). Here, we show that peptides ending with OMP-like C-terminal sequences bind the DegS PDZ domain, activate DegS cleavage of RseA, and induce sigma(E)-dependent transcription. These results suggest that DegS acts as a sensor of envelope stress by binding unassembled OMPs. DegS activation involves relief of inhibitory interactions between its PDZ and protease domains. Peptide binding to inhibitory PDZ domains in proteases related to DegS, including DegP/HtrA, may also regulate the degradation of specific substrates by these enzymes.


Assuntos
Proteínas de Bactérias/metabolismo , Escherichia coli/enzimologia , Proteínas de Membrana/metabolismo , Peptídeos/metabolismo , Porinas/metabolismo , Dobramento de Proteína , Proteínas de Bactérias/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Proteínas de Membrana/genética , Modelos Biológicos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Peptídeos/genética , Periplasma/enzimologia , Porinas/genética , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Fator sigma/genética , Fator sigma/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética
12.
Genes Dev ; 16(16): 2156-68, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12183369

RESUMO

All cells have stress response pathways that maintain homeostasis in each cellular compartment. In the Gram-negative bacterium Escherichia coli, the sigma(E) pathway responds to protein misfolding in the envelope. The stress signal is transduced across the inner membrane to the cytoplasm via the inner membrane protein RseA, the anti-sigma factor that inhibits the transcriptional activity of sigma(E). Stress-induced activation of the pathway requires the regulated proteolysis of RseA. In this report we show that RseA is degraded by sequential proteolytic events controlled by the inner membrane-anchored protease DegS and the membrane-embedded metalloprotease YaeL, an ortholog of mammalian Site-2 protease (S2P). This is consistent with the mechanism of activation of ATF6, the mammalian unfolded protein response transcription factor by Site-1 protease and S2P. Thus, mammalian and bacterial cells employ a conserved proteolytic mechanism to activate membrane-associated transcription factors that initiate intercompartmental cellular stress responses.


Assuntos
Proteínas de Bactérias/fisiologia , Endopeptidases/fisiologia , Proteínas de Escherichia coli/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Fator sigma/metabolismo , Fatores de Transcrição/metabolismo , Fator 6 Ativador da Transcrição , Bacillus subtilis/enzimologia , Bacillus subtilis/metabolismo , Sítios de Ligação , Western Blotting , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Escherichia coli/metabolismo , Mutação , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Espectrofotometria , Frações Subcelulares/metabolismo , Fatores de Tempo , Transdução Genética , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...