Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Genes (Basel) ; 14(1)2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36672963

RESUMO

The SOX transcription factor family is pivotal in controlling aspects of development. To identify genotype-phenotype relationships of SOX proteins, we performed a non-biased study of SOX using 1890 open-reading frame and 6667 amino acid sequences in combination with structural dynamics to interpret 3999 gnomAD, 485 ClinVar, 1174 Geno2MP, and 4313 COSMIC human variants. We identified, within the HMG (High Mobility Group)- box, twenty-seven amino acids with changes in multiple SOX proteins annotated to clinical pathologies. These sites were screened through Geno2MP medical phenotypes, revealing novel SOX15 R104G associated with musculature abnormality and SOX8 R159G with intellectual disability. Within gnomAD, SOX18 E137K (rs201931544), found within the HMG box of ~0.8% of Latinx individuals, is associated with seizures and neurological complications, potentially through blood-brain barrier alterations. A total of 56 highly conserved variants were found at sites outside the HMG-box, including several within the SOX2 HMG-box-flanking region with neurological associations, several in the SOX9 dimerization region associated with Campomelic Dysplasia, SOX14 K88R (rs199932938) flanking the HMG box associated with cardiovascular complications within European populations, and SOX7 A379V (rs143587868) within an SOXF conserved far C-terminal domain heterozygous in 0.716% of African individuals with associated eye phenotypes. This SOX data compilation builds a robust genotype-to-phenotype association for a gene family through more robust ortholog data integration.


Assuntos
Proteínas de Grupo de Alta Mobilidade , Fatores de Transcrição SOX , Humanos , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fatores de Transcrição SOX/genética , Sequência de Aminoácidos , Dimerização , Genótipo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Fatores de Transcrição SOXB2/genética , Fatores de Transcrição SOXB2/metabolismo , Fatores de Transcrição SOXE/genética
3.
Elife ; 102021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34142659

RESUMO

Caveolae-associated protein 3 (cavin3) is inactivated in most cancers. We characterized how cavin3 affects the cellular proteome using genome-edited cells together with label-free quantitative proteomics. These studies revealed a prominent role for cavin3 in DNA repair, with BRCA1 and BRCA1 A-complex components being downregulated on cavin3 deletion. Cellular and cell-free expression assays revealed a direct interaction between BRCA1 and cavin3 that occurs when cavin3 is released from caveolae that are disassembled in response to UV and mechanical stress. Overexpression and RNAi-depletion revealed that cavin3 sensitized various cancer cells to UV-induced apoptosis. Supporting a role in DNA repair, cavin3-deficient cells were sensitive to PARP inhibition, where concomitant depletion of 53BP1 restored BRCA1-dependent sensitivity to PARP inhibition. We conclude that cavin3 functions together with BRCA1 in multiple cancer-related pathways. The loss of cavin3 function may provide tumor cell survival by attenuating apoptotic sensitivity and hindering DNA repair under chronic stress conditions.


When cells become cancerous they often stop making certain proteins. This includes a protein known as cavin3 which resides in bulb-shaped pits of the membrane that surrounds the cell called caveolae. These structures work like stress detectors, picking up changes in the membrane and releasing proteins, such as cavin3, into the cell's interior. Past studies suggest that cavin3 might interact with a protein called BRCA1 that suppresses the formation of tumors. Cells with mutations in the gene for BRCA1 struggle to fix damage in their DNA, and have to rely on other repair proteins, such as PARPs (short for poly (ADP-ribose) polymerases). Blocking PARP proteins with drugs can kill cancer cells with problems in their BRCA1 proteins. However, it was unclear what role cavin3 plays in this mechanism. To investigate this, McMahon et al. exposed cells grown in the laboratory to DNA-damaging UV light to stimulate the release of cavin3 from caveolae. This revealed that cavin3 interacts with BRCA1 when cells are under stress, and helps stabilize the protein so it can perform DNA repairs. Cells without cavin3 showed decreased levels of the BRCA1 protein, but compensated for the loss of BRCA1 by increasing the levels of their PARP proteins. These cells also had increased DNA damage following treatment with drugs that block PARPs, similar to cancer cells carrying mutations in the gene for BRCA1. These findings suggest that cavin3 helps BRCA1 to suppress the formation of tumors, and therefore should be considered when developing new anti-cancer treatments.


Assuntos
Proteína BRCA1/metabolismo , Cavéolas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Estresse Fisiológico/genética , Apoptose/genética , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteoma/genética , Proteômica
4.
Emerg Microbes Infect ; 10(1): 178-195, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33372854

RESUMO

The genome of SARS-CoV-2 encodes two viral proteases (NSP3/papain-like protease and NSP5/3C-like protease) that are responsible for cleaving viral polyproteins during replication. Here, we discovered new functions of the NSP3 and NSP5 proteases of SARS-CoV-2, demonstrating that they could directly cleave proteins involved in the host innate immune response. We identified 3 proteins that were specifically and selectively cleaved by NSP3 or NSP5: IRF-3, and NLRP12 and TAB1, respectively. Direct cleavage of IRF3 by NSP3 could explain the blunted Type-I IFN response seen during SARS-CoV-2 infections while NSP5 mediated cleavage of NLRP12 and TAB1 point to a molecular mechanism for enhanced production of cytokines and inflammatory responThe genome of SARS-CoV-2 encodes two viral proteases (NSP3/papain-like protease and NSP5/3C-like protease) that are responsible for cleaving viral polyproteins during replication. Here, we discovered new functions of the NSP3 and NSP5 proteases of SARS-CoV-2, demonstrating that they could directly cleave proteins involved in the host innate immune response. We identified 3 proteins that were specifically and selectively cleaved by NSP3 or NSP5: IRF-3, and NLRP12 and TAB1, respectively. Direct cleavage of IRF3 by NSP3 could explain the blunted Type-I IFN response seen during SARS-CoV-2 infections while NSP5 mediated cleavage of NLRP12 and TAB1 point to a molecular mechanism for enhanced production of cytokines and inflammatory response observed in COVID-19 patients. We demonstrate that in the mouse NLRP12 protein, one of the recognition site is not cleaved in our in-vitro assay. We pushed this comparative alignment of IRF-3 and NLRP12 homologs and show that the lack or presence of cognate cleavage motifs in IRF-3 and NLRP12 could contribute to the presentation of disease in cats and tigers, for example. Our findings provide an explanatory framework for indepth studies into the pathophysiology of COVID-19.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteases 3C de Coronavírus/metabolismo , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sequência de Aminoácidos , Animais , COVID-19/patologia , Linhagem Celular , Quirópteros/virologia , Proteases 3C de Coronavírus/genética , Proteases Semelhantes à Papaína de Coronavírus/genética , Células HEK293 , Humanos , Camundongos , SARS-CoV-2/enzimologia , SARS-CoV-2/genética
5.
Nat Commun ; 10(1): 4288, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31537793

RESUMO

Macrophage-expressed gene 1 (MPEG1/Perforin-2) is a perforin-like protein that functions within the phagolysosome to damage engulfed microbes. MPEG1 is thought to form pores in target membranes, however, its mode of action remains unknown. We use cryo-Electron Microscopy (cryo-EM) to determine the 2.4 Å structure of a hexadecameric assembly of MPEG1 that displays the expected features of a soluble prepore complex. We further discover that MPEG1 prepore-like assemblies can be induced to perforate membranes through acidification, such as would occur within maturing phagolysosomes. We next solve the 3.6 Å cryo-EM structure of MPEG1 in complex with liposomes. These data reveal that a multi-vesicular body of 12 kDa (MVB12)-associated ß-prism (MABP) domain binds membranes such that the pore-forming machinery of MPEG1 is oriented away from the bound membrane. This unexpected mechanism of membrane interaction suggests that MPEG1 remains bound to the phagolysosome membrane while simultaneously forming pores in engulfed bacterial targets.


Assuntos
Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Bactérias/imunologia , Microscopia Crioeletrônica , Humanos , Lipossomos/metabolismo , Lisossomos/fisiologia , Macrófagos/imunologia , Microscopia de Força Atômica , Domínios Proteicos , Estrutura Secundária de Proteína
6.
Elife ; 82019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31358114

RESUMO

Propranolol is an approved non-selective ß-adrenergic blocker that is first line therapy for infantile hemangioma. Despite the clinical benefit of propranolol therapy in hemangioma, the mechanistic understanding of what drives this outcome is limited. Here, we report successful treatment of pericardial edema with propranolol in a patient with Hypotrichosis-Lymphedema-Telangiectasia and Renal (HLTRS) syndrome, caused by a mutation in SOX18. Using a mouse pre-clinical model of HLTRS, we show that propranolol treatment rescues its corneal neo-vascularisation phenotype. Dissection of the molecular mechanism identified the R(+)-propranolol enantiomer as a small molecule inhibitor of the SOX18 transcription factor, independent of any anti-adrenergic effect. Lastly, in a patient-derived in vitro model of infantile hemangioma and pre-clinical model of HLTRS we demonstrate the therapeutic potential of the R(+) enantiomer. Our work emphasizes the importance of SOX18 etiological role in vascular neoplasms, and suggests R(+)-propranolol repurposing to numerous indications ranging from vascular diseases to metastatic cancer.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Inibidores Enzimáticos/farmacologia , Hemangioma/tratamento farmacológico , Hipotricose/tratamento farmacológico , Linfedema/tratamento farmacológico , Propranolol/farmacologia , Fatores de Transcrição SOXF/antagonistas & inibidores , Telangiectasia/tratamento farmacológico , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Humanos , Camundongos , Modelos Teóricos , Propranolol/administração & dosagem
7.
Int J Mol Sci ; 20(6)2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30871132

RESUMO

Since their discovery in the early 20th century, antibiotics have been used as the primary weapon against bacterial infections. Due to their prophylactic effect, they are also used as part of the cocktail of drugs given to treat complex diseases such as cancer or during surgery, in order to prevent infection. This has resulted in a decrease of mortality from infectious diseases and an increase in life expectancy in the last 100 years. However, as a consequence of administering antibiotics broadly to the population and sometimes misusing them, antibiotic-resistant bacteria have appeared. The emergence of resistant strains is a global health threat to humanity. Highly-resistant bacteria like Staphylococcus aureus (methicillin-resistant) or Enterococcus faecium (vancomycin-resistant) have led to complications in intensive care units, increasing medical costs and putting patient lives at risk. The appearance of these resistant strains together with the difficulty in finding new antimicrobials has alarmed the scientific community. Most of the strategies currently employed to develop new antibiotics point towards novel approaches for drug design based on prodrugs or rational design of new molecules. However, targeting crucial bacterial processes by these means will keep creating evolutionary pressure towards drug resistance. In this review, we discuss antibiotic resistance and new options for antibiotic discovery, focusing in particular on new alternatives aiming to disarm the bacteria or empower the host to avoid disease onset.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Animais , Enterococcus faecium/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Humanos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Infecções Estafilocócicas/tratamento farmacológico
8.
EMBO Rep ; 20(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30498077

RESUMO

The murine cytomegalovirus protein M45 protects infected mouse cells from necroptotic death and, when heterologously expressed, can protect human cells from necroptosis induced by tumour necrosis factor receptor (TNFR) activation. Here, we show that the N-terminal 90 residues of the M45 protein, which contain a RIP homotypic interaction motif (RHIM), are sufficient to confer protection against TNFR-induced necroptosis. This N-terminal region of M45 drives rapid self-assembly into homo-oligomeric amyloid fibrils and interacts with the RHIMs of the human kinases RIPK1 and RIPK3, and the Z-DNA binding protein 1 (ZBP1), to form heteromeric amyloid fibrils in vitro Mutation of the tetrad residues in the M45 RHIM attenuates homo- and hetero-amyloid assembly by M45, suggesting that the amyloidogenic nature of the M45 RHIM underlies its biological activity. The M45 RHIM preferentially interacts with RIPK3 and ZBP1 over RIPK1 and alters the properties of the host RHIM protein assemblies. Our results indicate that M45 mimics the interactions made by RIPK1 or ZBP1 with RIPK3, thereby forming heteromeric amyloid structures, which may explain its ability to inhibit necroptosis.


Assuntos
Amiloide/metabolismo , Necroptose , Agregação Patológica de Proteínas/metabolismo , Multimerização Proteica , Ribonucleotídeo Redutases/metabolismo , Proteínas Virais/metabolismo , Amiloide/química , Amiloide/ultraestrutura , Amiloidose/etiologia , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Humanos , Camundongos , Modelos Moleculares , Ligação Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Ribonucleotídeo Redutases/química , Relação Estrutura-Atividade , Proteínas Virais/química
9.
BMC Biol ; 16(1): 149, 2018 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-30583727

RESUMO

BACKGROUND: Higher-order self-assembly of proteins, or "prion-like" polymerisation, is now emerging as a simple and robust mechanism for signal amplification, in particular within the innate immune system, where the recognition of pathogens or danger-associated molecular patterns needs to trigger a strong, binary response within cells. MyD88, an important adaptor protein downstream of TLRs, is one of the most recent candidates for involvement in signalling by higher order self-assembly. In this new light, we set out to re-interpret the role of polymerisation in MyD88-related diseases and study the impact of disease-associated point mutations L93P, R196C, and L252P/L265P at the molecular level. RESULTS: We first developed new in vitro strategies to characterise the behaviour of polymerising, full-length MyD88 at physiological levels. To this end, we used single-molecule fluorescence fluctuation spectroscopy coupled to a eukaryotic cell-free protein expression system. We were then able to explore the polymerisation propensity of full-length MyD88, at low protein concentration and without purification, and compare it to the behaviours of the isolated TIR domain and death domain that have been shown to have self-assembly properties on their own. These experiments demonstrate that the presence of both domains is required to cooperatively lead to efficient polymerisation of the protein. We then characterised three pathological mutants of MyD88. CONCLUSION: We discovered that all mutations block the ability of MyD88 to polymerise fully. Interestingly, we show that, in contrast to L93P and R196C, L252P is a gain-of-function mutation, which allows the MyD88 mutant to form extremely stable oligomers, even at low nanomolar concentrations. Thus, our results shed new light on the digital "all-or-none" responses by the myddosomes and the behaviour of the oncogenic mutations of MyD88.


Assuntos
Imunidade Inata/genética , Glicoproteínas de Membrana/genética , Mutação , Receptores de Interleucina-1/genética , Humanos , Sistema Imunitário/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Polimerização , Receptores de Interleucina-1/química , Receptores de Interleucina-1/metabolismo
10.
PLoS Biol ; 16(4): e2005473, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29621251

RESUMO

We describe the development and application of a suite of modular tools for high-resolution detection of proteins and intracellular protein complexes by electron microscopy (EM). Conditionally stable GFP- and mCherry-binding nanobodies (termed csGBP and csChBP, respectively) are characterized using a cell-free expression and analysis system and subsequently fused to an ascorbate peroxidase (APEX) enzyme. Expression of these cassettes alongside fluorescently labelled proteins results in recruitment and stabilisation of APEX, whereas unbound APEX nanobodies are efficiently degraded by the proteasome. This greatly simplifies correlative analyses, enables detection of less-abundant proteins, and eliminates the need to balance expression levels between fluorescently labelled and APEX nanobody proteins. Furthermore, we demonstrate the application of this system to bimolecular complementation ('EM split-fluorescent protein'), for localisation of protein-protein interactions at the ultrastructural level.


Assuntos
Ascorbato Peroxidases/genética , Células Epiteliais/ultraestrutura , Microscopia Eletrônica/métodos , Microscopia de Fluorescência/métodos , Imagem Individual de Molécula/métodos , Anticorpos de Domínio Único/química , Animais , Ascorbato Peroxidases/metabolismo , Linhagem Celular , Sistema Livre de Células , Cricetulus , Células Epiteliais/metabolismo , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/genética , Proteína Vermelha Fluorescente
11.
MAbs ; 10(4): 596-606, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29648920

RESUMO

Antibodies are routinely used to study the activity of transcription factors, using various in vitro and in vivo approaches such as electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, genome-wide method analysis coupled with next generation sequencing, or mass spectrometry. More recently, a new application for antibodies has emerged as crystallisation scaffolds for difficult to crystallise proteins, such as transcription factors. Only in a few rare cases, antibodies have been used to modulate the activity of transcription factors, and there is a real gap in our knowledge on how to efficiently design antibodies to interfere with transcription. The molecular function of transcription factors is underpinned by complex networks of protein-protein interaction and in theory, setting aside intra-cellular delivery challenges, developing antibody-based approaches to modulate transcription factor activity appears a viable option. Here, we demonstrate that antibodies or an antibody single-chain variable region fragments are powerful molecular tools to unravel complex protein-DNA and protein-protein binding mechanisms. In this study, we focus on the molecular mode of action of the transcription factor SOX18, a key modulator of endothelial cell fate during development, as well as an attractive target in certain pathophysiological conditions such as solid cancer metastasis. The engineered antibody we designed inhibits SOX18 transcriptional activity, by interfering specifically with an 8-amino-acid motif in the C-terminal region directly adjacent to α-Helix 3 of SOX18 HMG domain, thereby disrupting protein-protein interaction. This new approach establishes a framework to guide the study of transcription factors interactomes using antibodies as molecular handles.


Assuntos
Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/química , Anticorpos de Cadeia Única , Humanos
12.
Elife ; 62017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28137359

RESUMO

Pharmacological targeting of transcription factors holds great promise for the development of new therapeutics, but strategies based on blockade of DNA binding, nuclear shuttling, or individual protein partner recruitment have yielded limited success to date. Transcription factors typically engage in complex interaction networks, likely masking the effects of specifically inhibiting single protein-protein interactions. Here, we used a combination of genomic, proteomic and biophysical methods to discover a suite of protein-protein interactions involving the SOX18 transcription factor, a known regulator of vascular development and disease. We describe a small-molecule that is able to disrupt a discrete subset of SOX18-dependent interactions. This compound selectively suppressed SOX18 transcriptional outputs in vitro and interfered with vascular development in zebrafish larvae. In a mouse pre-clinical model of breast cancer, treatment with this inhibitor significantly improved survival by reducing tumour vascular density and metastatic spread. Our studies validate an interactome-based molecular strategy to interfere with transcription factor activity, for the development of novel disease therapeutics.


Assuntos
Antineoplásicos/metabolismo , Neoplasias da Mama/prevenção & controle , Fatores de Transcrição SOXF/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos , Animais , Fenômenos Biofísicos , Vasos Sanguíneos/embriologia , Modelos Animais de Doenças , Genômica , Camundongos , Proteômica , Resultado do Tratamento , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/antagonistas & inibidores
13.
J Cell Biol ; 214(6): 705-18, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27597756

RESUMO

Munc18-1 is a key component of the exocytic machinery that controls neurotransmitter release. Munc18-1 heterozygous mutations cause developmental defects and epileptic phenotypes, including infantile epileptic encephalopathy (EIEE), suggestive of a gain of pathological function. Here, we used single-molecule analysis, gene-edited cells, and neurons to demonstrate that Munc18-1 EIEE-causing mutants form large polymers that coaggregate wild-type Munc18-1 in vitro and in cells. Surprisingly, Munc18-1 EIEE mutants also form Lewy body-like structures that contain α-synuclein (α-Syn). We reveal that Munc18-1 binds α-Syn, and its EIEE mutants coaggregate α-Syn. Likewise, removal of endogenous Munc18-1 increases the aggregative propensity of α-Syn(WT) and that of the Parkinson's disease-causing α-Syn(A30P) mutant, an effect rescued by Munc18-1(WT) expression, indicative of chaperone activity. Coexpression of the α-Syn(A30P) mutant with Munc18-1 reduced the number of α-Syn(A30P) aggregates. Munc18-1 mutations and haploinsufficiency may therefore trigger a pathogenic gain of function through both the corruption of native Munc18-1 and a perturbed chaperone activity for α-Syn leading to aggregation-induced neurodegeneration.


Assuntos
Chaperonas Moleculares/metabolismo , Proteínas Munc18/metabolismo , Degeneração Neural , Neurônios/metabolismo , Agregados Proteicos , alfa-Sinucleína/metabolismo , Animais , Animais Recém-Nascidos , Genótipo , Haploinsuficiência , Corpos de Lewy/metabolismo , Corpos de Lewy/patologia , Microscopia de Fluorescência , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Proteínas Munc18/química , Proteínas Munc18/genética , Mutação , Neurônios/patologia , Células PC12 , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ligação Proteica , Conformação Proteica , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Transfecção , alfa-Sinucleína/química , alfa-Sinucleína/genética
14.
Proc Natl Acad Sci U S A ; 111(38): E3957-65, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25201979

RESUMO

Growth factor receptor levels are aberrantly high in diverse cancers, driving the proliferation and survival of tumor cells. Understanding the molecular basis for this aberrant elevation has profound clinical implications. Here we show that the pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) suppresses receptor tyrosine kinase (RTK) signaling output by a previously unidentified epigenetic mechanism unrelated to its previously described function as the hydrophobic motif phosphatase for the protein kinase AKT, protein kinase C, and S6 kinase. Specifically, we show that nuclear-localized PHLPP suppresses histone phosphorylation and acetylation, in turn suppressing the transcription of diverse growth factor receptors, including the EGF receptor. These data uncover a much broader role for PHLPP in regulation of growth factor signaling beyond its direct inactivation of AKT: By suppressing RTK levels, PHLPP dampens the downstream signaling output of two major oncogenic pathways, the PI3 kinase/AKT and the Rat sarcoma (RAS)/ERK pathways. Our data are consistent with a model in which PHLPP modifies the histone code to control the transcription of RTKs.


Assuntos
Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Modelos Biológicos , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Animais , Linhagem Celular Transformada , Receptores ErbB/genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas Fosfatases/genética , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Sequências Repetitivas de Aminoácidos , Transcrição Gênica/fisiologia
15.
Biochemistry ; 53(24): 3971-81, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24892992

RESUMO

PH domain leucine-rich repeat protein phosphatase (PHLPP) directly dephosphorylates and inactivates Akt and protein kinase C and is therefore a prime target for pharmacological intervention of two key signaling pathways, the phosphatidylinositol 3-kinase and diacylglycerol signaling pathways. Here we report on the first biochemical characterization of the phosphatase domain of a PHLPP family member. The human PHLPP1 and PHLPP2 phosphatase domains were expressed and purified from bacteria or insect cells and their activities compared to that of full-length proteins immunoprecipitated from mammalian cells. Biochemical analyses reveal that the PHLPP phosphatase domain effectively dephosphorylates synthetic and peptidic substrates, that its activity is modulated by metals and lipophilic compounds, and that it has relatively high thermal stability. Mutational analysis of PHLPP2 reveals an unusual active site architecture compared to the canonical architecture of PP2C phosphatases and identifies key acidic residues (Asp 806, Glu 989, and Asp 1024) and bulky aromatic residues (Phe 783 and Phe 808) whose mutation impairs activity. Consistent with a unique active site architecture, we identify inhibitors that discriminate between PHLPP2 and PP2Cα. These data establish PHLPP as a member of the PP2C family of phosphatases with a unique active site architecture.


Assuntos
Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Análise Mutacional de DNA , Estabilidade Enzimática , Humanos , Modelos Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/isolamento & purificação , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/isolamento & purificação , Fosforilação , Proteína Quinase C/metabolismo , Proteína Fosfatase 2C , Proteínas Proto-Oncogênicas c-akt/metabolismo
16.
Science ; 344(6185): 1249783, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24833397

RESUMO

Signaling from JAK (Janus kinase) protein kinases to STAT (signal transducers and activators of transcription) transcription factors is key to many aspects of biology and medicine, yet the mechanism by which cytokine receptors initiate signaling is enigmatic. We present a complete mechanistic model for activation of receptor-bound JAK2, based on an archetypal cytokine receptor, the growth hormone receptor. For this, we used fluorescence resonance energy transfer to monitor positioning of the JAK2 binding motif in the receptor dimer, substitution of the receptor extracellular domains with Jun zippers to control the position of its transmembrane (TM) helices, atomistic modeling of TM helix movements, and docking of the crystal structures of the JAK2 kinase and its inhibitory pseudokinase domain with an opposing kinase-pseudokinase domain pair. Activation of the receptor dimer induced a separation of its JAK2 binding motifs, driven by a ligand-induced transition from a parallel TM helix pair to a left-handed crossover arrangement. This separation leads to removal of the pseudokinase domain from the kinase domain of the partner JAK2 and pairing of the two kinase domains, facilitating trans-activation. This model may well generalize to other class I cytokine receptors.


Assuntos
Janus Quinase 2/metabolismo , Receptores da Somatotropina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Cisteína/química , Ativação Enzimática , Células HEK293 , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/química , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores da Somatotropina/química , Receptores da Somatotropina/genética
17.
Mol Cell ; 54(3): 378-91, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24703948

RESUMO

Akt activation is a hallmark of human cancers. Here, we report a critical mechanism for regulation of Akt activity by the splicing kinase SRPK1, a downstream Akt target for transducing growth signals to regulate splicing. Surprisingly, we find that SRPK1 has a tumor suppressor function because ablation of SRPK1 in mouse embryonic fibroblasts induces cell transformation. We link the phenotype to constitutive Akt activation from genome-wide phosphoproteomics analysis and discover that downregulated SRPK1 impairs the recruitment of the Akt phosphatase PHLPP1 (pleckstrin homology (PH) domain leucine-rich repeat protein phosphatase) to Akt. Interestingly, SRPK1 overexpression is also tumorigenic because excess SRPK1 squelches PHLPP1. Thus, aberrant SRPK1 expression in either direction induces constitutive Akt activation, providing a mechanistic basis for previous observations that SRPK1 is downregulated in some cancer contexts and upregulated in others.


Assuntos
Carcinogênese/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Adesão Celular , Células Cultivadas , Senescência Celular , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Ativação Enzimática , Feminino , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Nus , Transplante de Neoplasias , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carga Tumoral
18.
Methods Mol Biol ; 819: 561-73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22183558

RESUMO

We present an example-based description of virtual screening (VS) techniques used to identify new regulators of the Akt phosphatase PHLPP (PH domain Leucine repeat Protein Phosphatase). This enzyme opposes the effects of two kinases, Akt and PKC, which play a major role in cell growth and survival. Therefore, PHLPP is a potential therapeutic target in pathophysiologies where these pathways are either repressed, such as in diabetes and cardiovascular diseases, or over-activated as in cancer. To the best of our knowledge, no PHLPP inhibitors have been reported so far in the literature. In this study, we used a combination of chemical and virtual screening techniques that led to the identification of a number of inhibiting compounds with diverse scaffolds. These compounds bind PHLPP and inhibit cell death when tested in cellular assays. We employed GLIDE docking software to screen a library of more than 40,000 compounds selected from the NCI open depository (250,000 compounds) by similarity searches. We compare the efficiency at which we determined binding compounds from the chemical screen, and compare enrichment factors of the virtually discovered compounds over chemical screening.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Fosfoproteínas Fosfatases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interface Usuário-Computador , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Moleculares , Fosfoproteínas Fosfatases/química , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/química , Homologia Estrutural de Proteína
19.
J Med Chem ; 53(19): 6899-911, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20836557

RESUMO

PH domain Leucine-rich repeat protein phosphatase (PHLPP) directly dephosphorylates and inactivates Akt and protein kinase C, poising it as a prime target for pharmacological intervention of two major survival pathways. Here we report on the discovery of small molecule inhibitors of the phosphatase activity of PHLPP, a member of the PP2C family of phosphatases for which there are no general pharmacological inhibitors. First, the Diversity Set of the NCI was screened for inhibition of the purified phosphatase domain of PHLPP2 in vitro. Second, selected libraries from the open NCI database were docked into a virtual model of the phosphatase domain of PHLPP2, previously trained with our experimental data set, unveiling additional inhibitors. Biochemical and cellular assays resulted in the identification of two structurally diverse compounds that selectively inhibit PHLPP in vitro, increase Akt signaling in cells, and prevent apoptosis. Thus, chemical and virtual screening has resulted in the identification of small molecules that promote Akt signaling by inhibiting its negative regulator PHLPP.


Assuntos
Antraquinonas/síntese química , Compostos Azo/síntese química , Proteínas de Escherichia coli/antagonistas & inibidores , Fosfoproteínas Fosfatases/antagonistas & inibidores , Salicilatos/síntese química , Sequência de Aminoácidos , Antraquinonas/química , Antraquinonas/farmacologia , Apoptose/efeitos dos fármacos , Compostos Azo/química , Compostos Azo/farmacologia , Domínio Catalítico , Linhagem Celular Tumoral , Proteínas de Escherichia coli/química , Ensaios de Triagem em Larga Escala , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fosfoproteínas Fosfatases/química , Fosforilação , Proteínas Proto-Oncogênicas c-akt/agonistas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Salicilatos/química , Salicilatos/farmacologia , Homologia de Sequência de Aminoácidos , Bibliotecas de Moléculas Pequenas
20.
J Phys Chem B ; 114(10): 3559-66, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-20170092

RESUMO

A hydrophobic mismatch between protein length and membrane thickness can lead to a modification of protein conformation, function, and oligomerization. To study the role of hydrophobic mismatch, we have measured the change in mobility of transmembrane peptides possessing a hydrophobic helix of various length d(pi) in lipid membranes of giant vesicles. We also used a model system where the hydrophobic thickness of the bilayers, h, can be tuned at will. We precisely measured the diffusion coefficient of the embedded peptides and gained access to the apparent size of diffusing objects. For bilayers thinner than d(pi), the diffusion coefficient decreases, and the derived characteristic sizes are larger than the peptide radii. Previous studies suggest that peptides accommodate by tilting. This scenario was confirmed by ATR-FTIR spectroscopy. As the membrane thickness increases, the value of the diffusion coefficient increases to reach a maximum at h approximately = d(pi). We show that this variation in diffusion coefficient is consistent with a decrease in peptide tilt. To do so, we have derived a relation between the diffusion coefficient and the tilt angle, and we used this relation to derive the peptide tilt from our diffusion measurements. As the membrane thickness increases, the peptides raise (i.e., their tilt is reduced) and reach an upright position and a maximal mobility for h approximately = d(pi). Using accessibility measurements, we show that when the membrane becomes too thick, the peptide polar heads sink into the interfacial region. Surprisingly, this "pinching" behavior does not hinder the lateral diffusion of the transmembrane peptides. Ultimately, a break in the peptide transmembrane anchorage is observed and is revealed by a "jump" in the D values.


Assuntos
Bicamadas Lipídicas/química , Peptídeos/química , Interações Hidrofóbicas e Hidrofílicas , Fluidez de Membrana , Modelos Químicos , Modelos Moleculares , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA