Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biotechnol Prog ; 39(1): e3306, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36264017

RESUMO

Biologics manufacturing is capital and consumable intensive with need for advanced inventory planning to account for supply chain constraints. Early-stage process design and technology transfer are often challenging due to limited information on process variability regarding bioreactor titer, process yield, and product quality. Monte Carlo (MC) methods offer a stochastic modeling approach for process optimization where probabilities of occurrence for process inputs are incorporated into a deterministic model to simulate more likely scenarios for process outputs. In this study, we explore MC simulation-based design of a monoclonal antibody downstream manufacturing process. We demonstrate that this probabilistic approach offers more representative outcomes over the conventional worst-case approach where the theoretical minimum and maximum values of each process parameter are used without consideration for their probability of occurrence. Our work demonstrates case studies on more practically sizing unit operations to improve consumable utilization, thereby reducing manufacturing costs. We also used MC simulations to minimize process cadence by constraining the number of cycles per unit operation to fit facility preferences. By factoring in process uncertainty, we have implemented MC simulation-based facility fit analyses to efficiently plan for inventory when accounting for process constraints during technology transfer from lab-scale to clinical or commercial manufacturing.


Assuntos
Reatores Biológicos , Transferência de Tecnologia , Método de Monte Carlo , Simulação por Computador , Anticorpos Monoclonais
2.
J Biol Chem ; 297(3): 101079, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34391780

RESUMO

Phosphorylation (activation) and dephosphorylation (deactivation) of the slit diaphragm proteins NEPHRIN and NEPH1 are critical for maintaining the kidney epithelial podocyte actin cytoskeleton and, therefore, proper glomerular filtration. However, the mechanisms underlying these events remain largely unknown. Here we show that NEPHRIN and NEPH1 are novel receptor proteins for hepatocyte growth factor (HGF) and can be phosphorylated independently of the mesenchymal epithelial transition receptor in a ligand-dependent fashion through engagement of their extracellular domains by HGF. Furthermore, we demonstrate SH2 domain-containing protein tyrosine phosphatase-2-dependent dephosphorylation of these proteins. To establish HGF as a ligand, purified baculovirus-expressed NEPHRIN and NEPH1 recombinant proteins were used in surface plasma resonance binding experiments. We report high-affinity interactions of NEPHRIN and NEPH1 with HGF, although NEPHRIN binding was 20-fold higher than that of NEPH1. In addition, using molecular modeling we constructed peptides that were used to map specific HGF-binding regions in the extracellular domains of NEPHRIN and NEPH1. Finally, using an in vitro model of cultured podocytes and an ex vivo model of Drosophila nephrocytes, as well as chemically induced injury models, we demonstrated that HGF-induced phosphorylation of NEPHRIN and NEPH1 is centrally involved in podocyte repair. Taken together, this is the first study demonstrating a receptor-based function for NEPHRIN and NEPH1. This has important biological and clinical implications for the repair of injured podocytes and the maintenance of podocyte integrity.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Proteínas de Membrana/metabolismo , Animais , Linhagem Celular , Taxa de Filtração Glomerular/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Junções Intercelulares/metabolismo , Rim/patologia , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Camundongos , Peptídeos/metabolismo , Fosforilação , Podócitos/metabolismo , Ligação Proteica/fisiologia , Transdução de Sinais/fisiologia
3.
Sci Signal ; 8(376): ra46, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25969544

RESUMO

Complexes of signaling proteins that are nucleated upon activation of receptor tyrosine kinases are dynamic macromolecular assemblies held together by interactions, such as the recognition of phosphotyrosines by Src homology 2 (SH2) domains. We predicted that reversible binding and phosphatase activity enable dynamic regulation of these protein complexes, which could affect signal transduction. We explored how dynamics in the interactions among the epidermal growth factor (EGF) receptor (EGFR), GRB2-associated binder protein 1 (GAB1), and SH2 domain-containing phosphatase 2 (SHP2) affected EGFR signaling output, specifically SHP2 binding to tyrosine-phosphorylated GAB1, which relieves the autoinhibition of SHP2. Among the effects of activated SHP2 is increased extracellular signal-regulated kinase (ERK) activity. We found that in H1666 lung adenocarcinoma cells, EGFR-activated Src family kinases (SFKs) counteracted repeated GAB1 dephosphorylation events and maintained the association of SHP2 with phosphorylated GAB1 at a cytosolic site distal from EGFR. A computational model predicted that an experimentally verified delay in SFK inactivation after EGFR inactivation, combined with an amplification of GAB1 phosphorylation in cells with proteins in a specific range of concentrations, enabled GAB1 phosphorylation and GAB1-SHP2 complexes to persist longer than EGFR phosphorylation persisted in response to EGF. This SFK-dependent mechanism was specific to EGFR and did not occur in response to activation of the receptor tyrosine kinase c-MET. Thus, our results quantitatively describe a regulatory mechanism used by some receptor tyrosine kinases to remotely control the duration of a signal by regulating the persistence of a signaling protein complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptores ErbB/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Receptores ErbB/genética , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosforilação/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Quinases da Família src/genética
4.
J Cell Sci ; 127(Pt 16): 3555-67, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24951116

RESUMO

Information from multiple signaling axes is integrated in the determination of cellular phenotypes. Here, we demonstrate this aspect of cellular decision making in glioblastoma multiforme (GBM) cells by investigating the multivariate signaling regulatory functions of the protein tyrosine phosphatase SHP2 (also known as PTPN11). Specifically, we demonstrate that the ability of SHP2 to simultaneously drive ERK1/2 and antagonize STAT3 pathway activities produces qualitatively different effects on the phenotypes of proliferation and resistance to EGFR and c-MET co-inhibition. Whereas the ERK1/2 and STAT3 pathways independently promote proliferation and resistance to EGFR and c-MET co-inhibition, SHP2-driven ERK1/2 activity is dominant in driving cellular proliferation and SHP2-mediated antagonism of STAT3 phosphorylation prevails in the promotion of GBM cell death in response to EGFR and c-MET co-inhibition. Interestingly, the extent of these SHP2 signaling regulatory functions is diminished in glioblastoma cells that express sufficiently high levels of the EGFR variant III (EGFRvIII) mutant, which is commonly expressed in GBM. In cells and tumors that express EGFRvIII, SHP2 also antagonizes the phosphorylation of EGFRvIII and c-MET and drives expression of HIF-1α and HIF-2α, adding complexity to the evolving understanding of the regulatory functions of SHP2 in GBM.


Assuntos
Proliferação de Células , Glioblastoma/enzimologia , Sistema de Sinalização das MAP Quinases , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Feminino , Gefitinibe , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/fisiopatologia , Humanos , Indóis/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Quinazolinas/administração & dosagem , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Sulfonas/administração & dosagem
5.
Biophys J ; 102(9): 2012-21, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22824264

RESUMO

The tyrosine phosphorylated epidermal growth factor receptor (EGFR) initiates numerous cell signaling pathways. Although EGFR phosphorylation levels are ultimately determined by the balance of receptor kinase and protein tyrosine phosphatase (PTP) activities, the kinetics of EGFR dephosphorylation are not well understood. Previous models of EGFR signaling have generally neglected PTP activity or computed PTP activity by considering data that do not fully reveal the kinetics and compartmentalization of EGFR dephosphorylation. We developed a compartmentalized, mechanistic model to elucidate the kinetics of EGFR dephosphorylation and the coupling of this process to phosphorylation-dependent EGFR endocytosis. Model regression against data from HeLa cells for EGFR phosphorylation response to EGFR activation, PTP inhibition, and EGFR kinase inhibition led to the conclusion that EGFR dephosphorylation occurs at the plasma membrane and in the cell interior with a timescale that is smaller than that for ligand-mediated EGFR endocytosis. The model further predicted that sufficiently rapid dephosphorylation of EGFR at the plasma membrane could potentially impede EGFR endocytosis, consistent with recent experimental findings. Overall, our results suggest that PTPs regulate multiple receptor-level phenomena via their action at the plasma membrane and cell interior and point to new possibilities for targeting PTPs for modulation of EGFR dynamics.


Assuntos
Trifosfato de Adenosina/metabolismo , Membrana Celular/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Modelos Biológicos , Proteínas Tirosina Quinases/metabolismo , Animais , Simulação por Computador , Genes erbB-1 , Células HeLa , Humanos , Fosforilação , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...