Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38746415

RESUMO

Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEAD. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in spindle cell rhabdomyosarcoma. We demonstrate that, in contrast to VGLL2 and TEAD1, the fusion proteins are strong activators of TEAD-dependent transcription, and their function does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase p300 to control TEAD-mediated transcriptional and epigenetic landscapes. We showed that small molecule p300 inhibition can suppress fusion proteins-induced oncogenic transformation both in vitro and in vivo. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.

2.
Cancer Res ; 83(21): 3611-3623, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37603596

RESUMO

For a majority of patients with non-small cell lung cancer with EGFR mutations, treatment with EGFR inhibitors (EGFRi) induces a clinical response. Despite this initial reduction in tumor size, residual disease persists that leads to disease relapse. Elucidating the preexisting biological differences between sensitive cells and surviving drug-tolerant persister cells and deciphering how drug-tolerant cells evolve in response to treatment could help identify strategies to improve the efficacy of EGFRi. In this study, we tracked the origins and clonal evolution of drug-tolerant cells at a high resolution by using an expressed barcoding system coupled with single-cell RNA sequencing. This platform enabled longitudinal profiling of gene expression and drug sensitivity in response to EGFRi across a large number of clones. Drug-tolerant cells had higher expression of key survival pathways such as YAP and EMT at baseline and could also differentially adapt their gene expression following EGFRi treatment compared with sensitive cells. In addition, drug combinations targeting common downstream components (MAPK) or orthogonal factors (chemotherapy) showed greater efficacy than EGFRi alone, which is attributable to broader targeting of the heterogeneous EGFRi-tolerance mechanisms present in tumors. Overall, this approach facilitates thorough examination of clonal evolution in response to therapy that could inform the development of improved diagnostic approaches and treatment strategies for targeting drug-tolerant cells. SIGNIFICANCE: The evolution and heterogeneity of EGFR inhibitor tolerance are identified in a large number of clones at enhanced cellular and temporal resolution using an expressed barcode technology coupled with single-cell RNA sequencing.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Recidiva Local de Neoplasia , Tolerância a Medicamentos
3.
Nat Commun ; 13(1): 6744, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36347861

RESUMO

Targeting TEAD autopalmitoylation has been proposed as a therapeutic approach for YAP-dependent cancers. Here we show that TEAD palmitoylation inhibitor MGH-CP1 and analogues block cancer cell "stemness", organ overgrowth and tumor initiation in vitro and in vivo. MGH-CP1 sensitivity correlates significantly with YAP-dependency in a large panel of cancer cell lines. However, TEAD inhibition or YAP/TAZ knockdown leads to transient inhibition of cell cycle progression without inducing cell death, undermining their potential therapeutic utilities. We further reveal that TEAD inhibition or YAP/TAZ silencing leads to VGLL3-mediated transcriptional activation of SOX4/PI3K/AKT signaling axis, which contributes to cancer cell survival and confers therapeutic resistance to TEAD inhibitors. Consistently, combination of TEAD and AKT inhibitors exhibits strong synergy in inducing cancer cell death. Our work characterizes the therapeutic opportunities and limitations of TEAD palmitoylation inhibitors in cancers, and uncovers an intrinsic molecular mechanism, which confers potential therapeutic resistance.


Assuntos
Neoplasias , Fosfatidilinositol 3-Quinases , Humanos , Lipoilação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Transcrição SOXC/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição de Domínio TEA/metabolismo
4.
Cell Rep ; 40(3): 111125, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858546

RESUMO

PTEN and LKB1 are intimately associated with gastrointestinal tumorigenesis. Mutations of PTEN or LKB1 lead to Cowden syndrome and Peutz-Jeghers syndrome characterized by development of gastrointestinal polyps. However, the cells of origin of these polyps and underlying mechanism remain unclear. Here, we reveal that PTEN or LKB1 deficiency in Gli1+ gut mesenchymal cells, but not intestinal epithelium, drives polyp formation histologically resembling polyposis in human patients. Mechanistically, although PTEN and LKB1 converge to regulate mTOR/AKT signaling in various tumor contexts, we find that mTOR is essential for PTEN-deletion-induced polyp formation but is largely dispensable for polyposis induced by mesenchymal LKB1 deficiency. Altogether, our studies identify Gli1-expressing mesenchymal cells as a common cell of origin for polyposis associated with PTEN and LKB1 and reveal their engagement of different downstream pathways in gut mesenchyme to suppress gastrointestinal tumorigenesis.


Assuntos
Quinases Proteína-Quinases Ativadas por AMP/metabolismo , Neoplasias Colorretais , Síndrome de Peutz-Jeghers , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Humanos , PTEN Fosfo-Hidrolase/genética , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patologia , Proteínas Serina-Treonina Quinases/genética , Serina-Treonina Quinases TOR , Proteína GLI1 em Dedos de Zinco/genética
8.
Cell Stem Cell ; 26(5): 675-692.e8, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32259481

RESUMO

Intestinal homeostasis is tightly regulated by complex yet poorly understood signaling networks. Here, we demonstrate that Lats1/2, the core Hippo kinases, are essential to maintain Wnt pathway activity and intestinal stem cells. Lats1/2 deletion leads to loss of intestinal stem cells but drives Wnt-uncoupled crypt expansion. To explore the function of downstream transcriptional enhanced associate domain (TEAD) transcription factors, we identified a selective small-molecule reversible inhibitor of TEAD auto-palmitoylation that directly occupies its lipid-binding site and inhibits TEAD-mediated transcription in vivo. Combining this chemical tool with genetic and proteomics approaches, we show that intestinal Wnt inhibition by Lats deletion is Yes-associated protein (YAP)/transcriptional activator with PDZ-binding domain (TAZ) dependent but TEAD independent. Mechanistically, nuclear YAP/TAZ interact with Groucho/Transducin-Like Enhancer of Split (TLE) to block Wnt/T-cell factor (TCF)-mediated transcription, and dual inhibition of TEAD and Lats suppresses Wnt-uncoupled Myc upregulation and epithelial over-proliferation in Adenomatous polyposis coli (APC)-mutated intestine. Our studies highlight a pharmacological approach to inhibit TEAD palmitoylation and have important implications for targeting Wnt and Hippo signaling in human malignancies.


Assuntos
Neoplasias , Fatores de Transcrição , Humanos , Intestinos , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
9.
Cell Rep ; 29(10): 3200-3211.e4, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31801083

RESUMO

Uveal melanoma (UM), the most common ocular malignancy, is characterized by GNAQ/11 mutations. Hippo/YAP and Ras/mitogen-activated protein kinase (MAPK) emerge as two important signaling pathways downstream of G protein alpha subunits of the Q class (GαQ/11)-mediated transformation, although whether and how they contribute to UM genesis in vivo remain unclear. Here, we adapt an adeno-associated virus (AAV)-based ocular injection method to directly deliver Cre recombinase into the mouse uveal tract and demonstrate that Lats1/2 kinases suppress UM formation specifically in uveal melanocytes. We find that genetic activation of YAP, but not Kras, is sufficient to initiate UM. We show that YAP/TAZ activation induced by Lats1/2 deletion cooperates with Kras to promote UM progression via downstream transcriptional reinforcement. Furthermore, dual inhibition of YAP/TAZ and Ras/MAPK synergizes to suppress oncogenic growth of human UM cells. Our data highlight the functional significance of Lats-YAP/TAZ in UM initiation and progression in vivo and suggest combination inhibition of YAP/TAZ and Ras/MAPK as a new therapeutic strategy for UM.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/genética , Melanoma/genética , Melanoma/patologia , Transativadores/genética , Fatores de Transcrição/genética , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Feminino , Células HEK293 , Humanos , Melanócitos/patologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação/genética , Transdução de Sinais/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
10.
Dev Cell ; 43(1): 35-47.e4, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28943241

RESUMO

YAP/TAZ are the major mediators of mammalian Hippo signaling; however, their precise function in the gastrointestinal tract remains poorly understood. Here we dissect the distinct roles of YAP/TAZ in endodermal epithelium and mesenchyme and find that, although dispensable for gastrointestinal epithelial development and homeostasis, YAP/TAZ function as the critical molecular switch to coordinate growth and patterning in gut mesenchyme. Our genetic analyses reveal that Lats1/2 kinases suppress expansion of the primitive mesenchymal progenitors, where YAP activation also prevents induction of the smooth muscle lineage through transcriptional repression of Myocardin. During later development, zone-restricted downregulation of YAP/TAZ provides the positional cue and allows smooth muscle cell differentiation induced by Hedgehog signaling. Taken together, our studies identify the mesenchymal requirement of YAP/TAZ in the gastrointestinal tract and highlight the functional interplays between Hippo and Hedgehog signaling underlying temporal and spatial control of tissue growth and specification in developing gut.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/fisiologia , Epitélio/metabolismo , Trato Gastrointestinal/metabolismo , Proteínas Hedgehog/metabolismo , Mesoderma/metabolismo , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Aciltransferases , Animais , Proteínas de Ciclo Celular , Epitélio/patologia , Camundongos Transgênicos , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Proteínas de Sinalização YAP
11.
Cell Rep ; 14(5): 1169-1180, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26832411

RESUMO

The Tead family transcription factors are the major intracellular mediators of the Hippo-Yap pathway. Despite the importance of Hippo signaling in tumorigenesis, Tead-dependent downstream oncogenic programs and target genes in cancer cells remain poorly understood. Here, we characterize Tead4-mediated transcriptional networks in a diverse range of cancer cells, including neuroblastoma, colorectal, lung, and endometrial carcinomas. By intersecting genome-wide chromatin occupancy analyses of Tead4, JunD, and Fra1/2, we find that Tead4 cooperates with AP1 transcription factors to coordinate target gene transcription. We find that Tead-AP1 interaction is JNK independent but engages the SRC1-3 co-activators to promote downstream transcription. Furthermore, we show that Tead-AP1 cooperation regulates the activity of the Dock-Rac/CDC42 module and drives the expression of a unique core set of target genes, thereby directing cell migration and invasion. Together, our data unveil a critical regulatory mechanism underlying Tead- and AP1-controlled transcriptional and functional outputs in cancer cells.


Assuntos
Movimento Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas Musculares/metabolismo , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Bases , Linhagem Celular Tumoral , Análise por Conglomerados , Genoma Humano , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/patologia , Ligação Proteica , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição de Domínio TEA
12.
Dev Cell ; 31(3): 291-304, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25453828

RESUMO

Similar to the mammalian intestine, the Drosophila adult midgut has resident stem cells that support growth and regeneration. How the niche regulates intestinal stem cell activity in both mammals and flies is not well understood. Here, we show that the conserved germinal center protein kinase Misshapen restricts intestinal stem cell division by repressing the expression of the JAK-STAT pathway ligand Upd3 in differentiating enteroblasts. Misshapen, a distant relative to the prototypic Warts activating kinase Hippo, interacts with and activates Warts to negatively regulate the activity of Yorkie and the expression of Upd3. The mammalian Misshapen homolog MAP4K4 similarly interacts with LATS (Warts homolog) and promotes inhibition of YAP (Yorkie homolog). Together, this work reveals that the Misshapen-Warts-Yorkie pathway acts in enteroblasts to control niche signaling to intestinal stem cells. These findings also provide a model in which to study requirements for MAP4K4-related kinases in MST1/2-independent regulation of LATS and YAP.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Animais , Diferenciação Celular/fisiologia , Divisão Celular , Regeneração/genética , Células-Tronco/citologia , Proteínas de Sinalização YAP
13.
Mol Cell ; 51(2): 211-25, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23769673

RESUMO

Dysregulation of Wnt signaling is closely associated with human liver tumorigenesis. However, liver cancer-specific Wnt transcriptional programs and downstream effectors remain poorly understood. Here, we identify tribbles homolog 2 (TRIB2) as a direct target of Wnt/TCF in liver cancer and demonstrate that transcription of Wnt target genes, including TRIB2, is coordinated by the TCF and FoxA transcription factors in liver cancer cells. We show that Wnt-TRIB2 activation is critical for cancer cell survival and transformation. Mechanistically, TRIB2 promotes protein stabilization of the YAP transcription coactivator through interaction with the ßTrCP ubiquitin ligase. Furthermore, we find that TRIB2 relieves the liver tumor suppressor protein C/EBPα-mediated inhibition of YAP/TEAD transcriptional activation in liver cancer cells. Altogether, our study uncovers a regulatory mechanism underlying liver cancer-specific Wnt transcriptional output, and suggests that TRIB2 functions as a signaling nexus to integrate the Wnt/ß-catenin, Hippo/YAP, and C/EBPα pathways in cancer cells.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Nucleares/metabolismo , Fator 1 de Transcrição de Linfócitos T/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator 1 de Transcrição de Linfócitos T/genética , Fatores de Transcrição/genética , Proteínas Wnt/genética , beta Catenina/genética
14.
J Biol Chem ; 288(24): 17589-96, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23645682

RESUMO

Hedgehog (Hh) signaling is involved in multiple aspects of embryonic gut development, including mesenchymal growth and smooth muscle differentiation. The Gli family transcription factors is thought to collectively mediate Hh signaling in mammals. However, the function of different Gli proteins in gut development remains uncharacterized. Here, we genetically dissect the contribution of Gli transcriptional activation and de-repression in intestinal growth and patterning. We find that removal of the Gli3 repressor is dispensable for intestinal development and does not play a major role in Hh-controlled gut development. However, Gli2 activation is able to fully rescue the Smoothened (Smo)-null intestinal phenotype, suggesting that the Gli2 transcription factor is the main effector for Hh signaling in the intestine. To understand further the molecular mechanism underlying Hh/Gli function in the developing gut, we identify a subset of small leucine-rich glycoproteins (SLRPs) that may function downstream of Hh signaling in the mesenchyme. We show that osteoglycin, a SLRP, inhibits Hh-induced differentiation toward the smooth muscle lineage in C3H10T1/2 pluripotent mesenchymal cells. Taken together, our study reveals, for the first time, the distinct roles of Gli proteins in intestine development and suggests SLRPs as novel regulators of smooth muscle cell differentiation.


Assuntos
Intestinos/embriologia , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Intestinos/citologia , Mesoderma/embriologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Miócitos de Músculo Liso/fisiologia , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Receptor Smoothened , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco
15.
Proc Natl Acad Sci U S A ; 109(17): E1038-47, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22493246

RESUMO

Pancreatic ductal adenocarcinoma (PDAC), one of the most aggressive human malignancies, is thought to be initiated by KRAS activation. Here we find that transcriptional activation mediated by the Gli family of transcription factors, although dispensable for pancreatic development, is required for Kras-induced proliferation and survival in primary pancreatic epithelial cells in culture and for Kras-driven pancreatic intraepithelial neoplasia and PDAC formation in vivo. Further, ectopic Gli1 activation in the mouse pancreas accelerates Kras-driven tumor formation, underscoring the importance of Gli transcription factors in pancreatic tumorigenesis. Interestingly, we demonstrate Gli-regulated I-kappa-B kinase epsilon (IKBKE) and NF-κB activity in pancreatic cancer cells and show that this activity is a critical downstream mediator for Gli-dependent PDAC cell transformation and survival. Together, these studies demonstrate the requirement for Gli in Kras-dependent pancreatic epithelial transformation, suggest a mechanism of Gli-NF-κB oncogenic activation, and provide genetic evidence supporting the therapeutic targeting of Gli activity in pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/genética , Genes ras , Neoplasias Pancreáticas/genética , Fatores de Transcrição/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Proteínas I-kappa B/metabolismo , Camundongos , NF-kappa B/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/metabolismo
16.
Methods Mol Biol ; 821: 329-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22125076

RESUMO

Extensive efforts are underway to develop small-molecule inhibitors of the mammalian target of rapamycin (mTOR) kinase. It is hoped that these inhibitors will have widespread clinical impact in oncology because mTOR is a major downstream effector of PI3K signaling, one of the most frequently activated pathways in cancer. In cells, mTOR is the catalytic core subunit of two distinct complexes, mTORC1 and mTORC2, which are defined by unique mTOR-interacting proteins and have unique functions downstream of PI3K. Two classes of mTOR inhibitors are currently being evaluated as cancer therapeutics: rapamycin and its analogs, which partially inhibit mTORC1 and in some cell types mTORC2, and the recently described ATP-competitive inhibitors, which inhibit the kinase activity of both complexes. Although small molecules that selectively target mTORC2 do not yet exist, experiments using mouse genetics suggest that a theoretical mTORC2 inhibitor may have significant therapeutic value. Here, we discuss an approach to model mTOR complex specific inhibitors using mouse genetics and how it can be applied to other gene products involved in oncogenic signaling to which inhibitors do not exist.


Assuntos
Técnicas de Inativação de Genes , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Transativadores/antagonistas & inibidores , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Proliferação de Células , Modelos Animais de Doenças , Descoberta de Drogas , Humanos , Masculino , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/genética , Transdução de Sinais/genética , Sirolimo/análogos & derivados , Fatores de Transcrição
17.
Biochemistry ; 48(5): 1025-35, 2009 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19138128

RESUMO

Siderophores are key virulence factors that allow bacteria to grow in iron-restricted environments. The Gram-positive pathogen Staphylococcus aureus is known to produce four siderophores for which genetic and/or structural data are unknown. Here we characterize the gene cluster responsible for producing the prevalent siderophore staphyloferrin A. In addition to expressing the cluster in the heterologous host Escherichia coli, which confers the ability to synthesize the siderophore, we reconstituted staphyloferrin A biosynthesis in vitro by expressing and purifying two key enzymes in the pathway. As with other polycarboxylate siderophores, staphyloferrin A is biosynthesized using the recently described nonribosomal peptide synthetase independent siderophore (NIS) biosynthetic pathway. Two NIS synthetases condense two molecules of citric acid to d-ornithine in a stepwise ordered process with SfnaD using the delta-amine as a nucleophile to form the first amide followed by SfnaB utilizing the alpha-amine to complete staphyloferrin A synthesis.


Assuntos
Proteínas de Bactérias/genética , Família Multigênica , Ornitina/análogos & derivados , Peptídeo Sintases/genética , Sideróforos/genética , Staphylococcus aureus/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/química , Citratos/química , Biologia Computacional/métodos , Escherichia coli/enzimologia , Escherichia coli/genética , Código Genético , Ornitina/química , Ornitina/genética , Peptídeo Sintases/química , Sideróforos/biossíntese , Sideróforos/química , Staphylococcus aureus/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...