Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 625(7995): 585-592, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38200309

RESUMEN

Oncogene-induced replication stress generates endogenous DNA damage that activates cGAS-STING-mediated signalling and tumour suppression1-3. However, the precise mechanism of cGAS activation by endogenous DNA damage remains enigmatic, particularly given that high-affinity histone acidic patch (AP) binding constitutively inhibits cGAS by sterically hindering its activation by double-stranded DNA (dsDNA)4-10. Here we report that the DNA double-strand break sensor MRE11 suppresses mammary tumorigenesis through a pivotal role in regulating cGAS activation. We demonstrate that binding of the MRE11-RAD50-NBN complex to nucleosome fragments is necessary to displace cGAS from acidic-patch-mediated sequestration, which enables its mobilization and activation by dsDNA. MRE11 is therefore essential for cGAS activation in response to oncogenic stress, cytosolic dsDNA and ionizing radiation. Furthermore, MRE11-dependent cGAS activation promotes ZBP1-RIPK3-MLKL-mediated necroptosis, which is essential to suppress oncogenic proliferation and breast tumorigenesis. Notably, downregulation of ZBP1 in human triple-negative breast cancer is associated with increased genome instability, immune suppression and poor patient prognosis. These findings establish MRE11 as a crucial mediator that links DNA damage and cGAS activation, resulting in tumour suppression through ZBP1-dependent necroptosis.


Asunto(s)
Transformación Celular Neoplásica , Proteína Homóloga de MRE11 , Nucleosomas , Nucleotidiltransferasas , Humanos , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Daño del ADN , Proteína Homóloga de MRE11/metabolismo , Necroptosis , Nucleosomas/metabolismo , Nucleotidiltransferasas/metabolismo , Radiación Ionizante , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Inestabilidad Genómica
2.
Mol Cell ; 62(6): 929-942, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27237051

RESUMEN

The retinoblastoma (Rb) protein exerts its tumor suppressor function primarily by inhibiting the E2F family of transcription factors that govern cell-cycle progression. However, it remains largely elusive whether the hyper-phosphorylated, non-E2F1-interacting form of Rb has any physiological role. Here we report that hyper-phosphorylated Rb directly binds to and suppresses the function of mTORC2 but not mTORC1. Mechanistically, Rb, but not p107 or p130, interacts with Sin1 and blocks the access of Akt to mTORC2, leading to attenuated Akt activation and increased sensitivity to chemotherapeutic drugs. As such, inhibition of Rb phosphorylation by depleting cyclin D or using CDK4/6 inhibitors releases Rb-mediated mTORC2 suppression. This, in turn, leads to elevated Akt activation to confer resistance to chemotherapeutic drugs in Rb-proficient cells, which can be attenuated with Akt inhibitors. Therefore, our work provides a molecular basis for the synergistic usage of CDK4/6 and Akt inhibitors in treating Rb-proficient cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Complejos Multiproteicos/metabolismo , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Ciclina D/genética , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Activación Enzimática , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
3.
Cell Mol Life Sci ; 80(6): 149, 2023 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-37183204

RESUMEN

STING acts as a cytosolic nucleotide sensor to trigger host defense upon viral or bacterial infection. While STING hyperactivation can exert anti-tumor effects by increasing T cell filtrates, in other contexts hyperactivation of STING can contribute to autoimmune and neuroinflammatory diseases. Several STING targeting agonists and a smaller subset of antagonists have been developed, yet STING targeted degraders, or PROTACs, remain largely underexplored. Here, we report a series of STING-agonist derived PROTACs that promote STING degradation in renal cell carcinoma (RCC) cells. We show that our STING PROTACs activate STING and target activated/phospho-STING for degradation. Locking STING on the endoplasmic reticulum via site-directed mutagenesis disables STING translocation to the proteasome and resultingly blocks STING degradation. We also demonstrate that PROTAC treatment blocks downstream innate immune signaling events and attenuates the anti-viral response. Interestingly, we find that VHL acts as a bona fide E3 ligase for STING in RCC; thus, VHL-recruiting STING PROTACs further promote VHL-dependent STING degradation. Our study reveals the design and biological assessment of VHL-recruiting agonist-derived STING PROTACs, as well as demonstrates an example of hijacking a physiological E3 ligase to enhance target protein degradation via distinct mechanisms.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Quimera Dirigida a la Proteólisis , Carcinoma de Células Renales/tratamiento farmacológico , Ubiquitina-Proteína Ligasas/metabolismo , Proteolisis , Neoplasias Renales/tratamiento farmacológico , Inmunidad Innata , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
4.
Semin Cancer Biol ; 85: 69-94, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34175443

RESUMEN

Cancer is the second leading cause of human death globally. PI3K/Akt/mTOR signaling is one of the most frequently dysregulated signaling pathways observed in cancer patients that plays crucial roles in promoting tumor initiation, progression and therapy responses. This is largely due to that PI3K/Akt/mTOR signaling is indispensable for many cellular biological processes, including cell growth, metastasis, survival, metabolism, and others. As such, small molecule inhibitors targeting major kinase components of the PI3K/Akt/mTOR signaling pathway have drawn extensive attention and been developed and evaluated in preclinical models and clinical trials. Targeting a single kinase component within this signaling usually causes growth arrest rather than apoptosis associated with toxicity-induced adverse effects in patients. Combination therapies including PI3K/Akt/mTOR inhibitors show improved patient response and clinical outcome, albeit developed resistance has been reported. In this review, we focus on revealing the mechanisms leading to the hyperactivation of PI3K/Akt/mTOR signaling in cancer and summarizing efforts for developing PI3K/Akt/mTOR inhibitors as either mono-therapy or combination therapy in different cancer settings. We hope that this review will facilitate further understanding of the regulatory mechanisms governing dysregulation of PI3K/Akt/mTOR oncogenic signaling in cancer and provide insights into possible future directions for targeted therapeutic regimen for cancer treatment, by developing new agents, drug delivery systems, or combination regimen to target the PI3K/Akt/mTOR signaling pathway. This information will also provide effective patient stratification strategy to improve the patient response and clinical outcome for cancer patients with deregulated PI3K/Akt/mTOR signaling.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-akt , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Terapia Molecular Dirigida , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Neoplasias/tratamiento farmacológico , Neoplasias/genética
5.
Nature ; 545(7654): 365-369, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28489822

RESUMEN

The mechanistic target of rapamycin (mTOR) has a key role in the integration of various physiological stimuli to regulate several cell growth and metabolic pathways. mTOR primarily functions as a catalytic subunit in two structurally related but functionally distinct multi-component kinase complexes, mTOR complex 1 (mTORC1) and mTORC2 (refs 1, 2). Dysregulation of mTOR signalling is associated with a variety of human diseases, including metabolic disorders and cancer. Thus, both mTORC1 and mTORC2 kinase activity is tightly controlled in cells. mTORC1 is activated by both nutrients and growth factors, whereas mTORC2 responds primarily to extracellular cues such as growth-factor-triggered activation of PI3K signalling. Although both mTOR and GßL (also known as MLST8) assemble into mTORC1 and mTORC2 (refs 11, 12, 13, 14, 15), it remains largely unclear what drives the dynamic assembly of these two functionally distinct complexes. Here we show, in humans and mice, that the K63-linked polyubiquitination status of GßL dictates the homeostasis of mTORC2 formation and activation. Mechanistically, the TRAF2 E3 ubiquitin ligase promotes K63-linked polyubiquitination of GßL, which disrupts its interaction with the unique mTORC2 component SIN1 (refs 12, 13, 14) to favour mTORC1 formation. By contrast, the OTUD7B deubiquitinase removes polyubiquitin chains from GßL to promote GßL interaction with SIN1, facilitating mTORC2 formation in response to various growth signals. Moreover, loss of critical ubiquitination residues in GßL, by either K305R/K313R mutations or a melanoma-associated GßL(ΔW297) truncation, leads to elevated mTORC2 formation, which facilitates tumorigenesis, in part by activating AKT oncogenic signalling. In support of a physiologically pivotal role for OTUD7B in the activation of mTORC2/AKT signalling, genetic deletion of Otud7b in mice suppresses Akt activation and Kras-driven lung tumorigenesis in vivo. Collectively, our study reveals a GßL-ubiquitination-dependent switch that fine-tunes the dynamic organization and activation of the mTORC2 kinase under both physiological and pathological conditions.


Asunto(s)
Carcinogénesis , Endopeptidasas/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Endopeptidasas/deficiencia , Endopeptidasas/genética , Activación Enzimática , Femenino , Homeostasis , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Complejos Multiproteicos/biosíntesis , Complejos Multiproteicos/química , Fosforilación , Poliubiquitina/metabolismo , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/química , Homóloga LST8 de la Proteína Asociada al mTOR
6.
Mol Cell ; 59(6): 917-30, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26344095

RESUMEN

The ERG gene is fused to TMPRSS2 in approximately 50% of prostate cancers (PrCa), resulting in its overexpression. However, whether this is the sole mechanism underlying ERG elevation in PrCa is currently unclear. Here we report that ERG ubiquitination and degradation are governed by the Cullin 3-based ubiquitin ligase SPOP and that deficiency in this pathway leads to aberrant elevation of the ERG oncoprotein. Specifically, we find that truncated ERG (ΔERG), encoded by the ERG fusion gene, is stabilized by evading SPOP-mediated destruction, whereas prostate cancer-associated SPOP mutants are also deficient in promoting ERG ubiquitination. Furthermore, we show that the SPOP/ERG interaction is modulated by CKI-mediated phosphorylation. Importantly, we demonstrate that DNA damage drugs, topoisomerase inhibitors, can trigger CKI activation to restore the SPOP/ΔERG interaction and its consequent degradation. Therefore, SPOP functions as a tumor suppressor to negatively regulate the stability of the ERG oncoprotein in prostate cancer.


Asunto(s)
Proteínas Nucleares/fisiología , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/fisiología , Transactivadores/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Antineoplásicos Fitogénicos/farmacología , Línea Celular Tumoral , Movimiento Celular , Proteínas Cullin/metabolismo , Progresión de la Enfermedad , Etopósido/farmacología , Células HEK293 , Humanos , Masculino , Datos de Secuencia Molecular , Invasividad Neoplásica , Neoplasias de la Próstata/patología , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Regulador Transcripcional ERG , Proteínas Supresoras de Tumor/fisiología
7.
Mol Cell ; 57(4): 648-661, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25661488

RESUMEN

Deficiency in repair of damaged DNA leads to genomic instability and is closely associated with tumorigenesis. Most DNA double-strand-breaks (DSBs) are repaired by two major mechanisms, homologous-recombination (HR) and non-homologous-end-joining (NHEJ). Although Akt has been reported to suppress HR, its role in NHEJ remains elusive. Here, we report that Akt phosphorylates XLF at Thr181 to trigger its dissociation from the DNA ligase IV/XRCC4 complex, and promotes its interaction with 14-3-3ß leading to XLF cytoplasmic retention, where cytosolic XLF is subsequently degraded by SCF(ß-TRCP) in a CKI-dependent manner. Physiologically, upon DNA damage, XLF-T181E expressing cells display impaired NHEJ and elevated cell death. Whereas a cancer-patient-derived XLF-R178Q mutant, deficient in XLF-T181 phosphorylation, exhibits an elevated tolerance of DNA damage. Together, our results reveal a pivotal role for Akt in suppressing NHEJ and highlight the tight connection between aberrant Akt hyper-activation and deficiency in timely DSB repair, leading to genomic instability and tumorigenesis.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Enzimas Reparadoras del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Carcinogénesis/genética , Citoplasma/metabolismo , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Humanos , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Ligasas SKP Cullina F-box/fisiología , Alineación de Secuencia
8.
Inorg Chem ; 61(40): 16009-16019, 2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-36153966

RESUMEN

The functionalized design of metal-organic frameworks (MOFs) has been rapidly developed in the last 20 years, and its broad applicability has been demonstrated in many fields. MOFs with desired functions can be assembled using predesigned organic linkers with specific metal nodes, which possess the ordered functional sites and open structures. Although a large number of carboxylic acid junctions have been used to construct MOFs, it is still a great challenge to realize their multifunctionality. In particular, there is a relative lack of research on MOFs as direct photocatalysts, which require not only abundant active sites and open structures but also adsorption groups and effective electron-hole separation performance. To this end, MOFs constructed from the carboxylic acid ligands derived from lophine-based derivatives and copper ions were deliberately used as a photocatalyst, and then, their application in dye degradation and aromatic alcohol conversion was investigated. In addition, in combination with the abundant Lewis sites of copper ions and imidazole sites, the material shows not only the adsorption and separation of C2 series and dyes but also the application of dye degradation and conversion of aromatic alcohols under illumination conditions. The corresponding results fully illustrate that the MOF constructed by using lophine derivatives can be an effective way to prepare photocatalysts. The subsequent research ideas will focus on designing a series of MOFs constructed with multilinked moieties of lophine groups and exploring their application strategies in the field of photocatalysis.

9.
Curr Issues Mol Biol ; 35: 59-84, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31422933

RESUMEN

Ubiquitin and ubiquitin-like modifiers, such as SUMO, exert distinct physiological functions by conjugating to protein substrates. Ubiquitination or SUMOylation of protein substrates determine the fate of modified proteins, including proteasomal degradation, cellular re-localization, alternations in binding partners and serving as a protein-binding platform, in a ubiquitin or SUMO linkage-dependent manner. DNA damage occurs constantly in living organisms but is also repaired by distinct tightly controlled mechanisms including homologous recombination, non-homologous end joining, inter-strand crosslink repair, nucleotide excision repair and base excision repair. On sensing damaged DNA, a ubiquitination/SUMOylation landscape is established to recruit DNA damage repair factors. Meanwhile, misloaded and mission-completed repair factors will be turned over by ubiquitin or SUMO modifications as well. These ubiquitination and SUMOylation events are tightly controlled by both E3 ubiquitin/SUMO ligases and deubiquitinases/deSUMOylases. In this review, we will summarize identified ubiquitin and SUMO-related modifications and their function in distinct DNA damage repair pathways, and provide evidence for responsible E3 ligases, deubiquitinases, SUMOylases and deSUMOylases in these processes. Given that genome instability leads to human disorders including cancer, understanding detailed molecular mechanisms for ubiquitin and SUMO-related regulations in DNA damage response may provide novel insights into therapeutic modalities to treat human diseases associated with deregulated DNA damage response.


Asunto(s)
Daño del ADN , Reparación del ADN , Transducción de Señal/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Reparación del ADN por Unión de Extremidades/genética , Recombinación Homóloga/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética
10.
Nature ; 508(7497): 541-5, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24670654

RESUMEN

Akt, also known as protein kinase B, plays key roles in cell proliferation, survival and metabolism. Akt hyperactivation contributes to many pathophysiological conditions, including human cancers, and is closely associated with poor prognosis and chemo- or radiotherapeutic resistance. Phosphorylation of Akt at S473 (ref. 5) and T308 (ref. 6) activates Akt. However, it remains unclear whether further mechanisms account for full Akt activation, and whether Akt hyperactivation is linked to misregulated cell cycle progression, another cancer hallmark. Here we report that Akt activity fluctuates across the cell cycle, mirroring cyclin A expression. Mechanistically, phosphorylation of S477 and T479 at the Akt extreme carboxy terminus by cyclin-dependent kinase 2 (Cdk2)/cyclin A or mTORC2, under distinct physiological conditions, promotes Akt activation through facilitating, or functionally compensating for, S473 phosphorylation. Furthermore, deletion of the cyclin A2 allele in the mouse olfactory bulb leads to reduced S477/T479 phosphorylation and elevated cellular apoptosis. Notably, cyclin A2-deletion-induced cellular apoptosis in mouse embryonic stem cells is partly rescued by S477D/T479E-Akt1, supporting a physiological role for cyclin A2 in governing Akt activation. Together, the results of our study show Akt S477/T479 phosphorylation to be an essential layer of the Akt activation mechanism to regulate its physiological functions, thereby providing a new mechanistic link between aberrant cell cycle progression and Akt hyperactivation in cancer.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/genética , Proliferación Celular , Ciclina A2/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Activación Enzimática , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Bulbo Olfatorio/citología , Bulbo Olfatorio/enzimología , Bulbo Olfatorio/metabolismo , Proteína Oncogénica v-akt/química , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Fosfoserina/metabolismo , Fosfotreonina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
11.
Mol Cell ; 44(2): 290-303, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-22017875

RESUMEN

The activities of both mTORC1 and mTORC2 are negatively regulated by their endogenous inhibitor, DEPTOR. As such, the abundance of DEPTOR is a critical determinant in the activity status of the mTOR network. DEPTOR stability is governed by the 26S-proteasome through a largely unknown mechanism. Here we describe an mTOR-dependent phosphorylation-driven pathway for DEPTOR destruction via SCF(ßTrCP). DEPTOR phosphorylation by mTOR in response to growth signals, and in collaboration with casein kinase I (CKI), generates a phosphodegron that binds ßTrCP. Failure to degrade DEPTOR through either degron mutation or ßTrCP depletion leads to reduced mTOR activity, reduced S6 kinase activity, and activation of autophagy to reduce cell growth. This work expands the current understanding of mTOR regulation by revealing a positive feedback loop involving mTOR and CKI-dependent turnover of its inhibitor, DEPTOR, suggesting that misregulation of the DEPTOR destruction pathway might contribute to aberrant activation of mTOR in disease.


Asunto(s)
Proteínas Ligasas SKP Cullina F-box/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Autofagia , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fosforilación , Transducción de Señal , Transfección
12.
Genes Dev ; 24(20): 2303-16, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20952539

RESUMEN

Drosophila contains one (dCDK12) and humans contain two (hCDK12 and hCDK13) proteins that are the closest evolutionary relatives of yeast Ctk1, the catalytic subunit of the major elongation-phase C-terminal repeat domain (CTD) kinase in Saccharomyces cerevisiae, CTDK-I. However, until now, neither CDK12 nor CDK13 has been demonstrated to be a bona fide CTD kinase. Using Drosophila, we demonstrate that dCDK12 (CG7597) is a transcription-associated CTD kinase, the ortholog of yCtk1. Fluorescence microscopy reveals that the distribution of dCDK12 on formaldehyde-fixed polytene chromosomes is virtually identical to that of hyperphosphorylated RNA polymerase II (RNAPII), but is distinct from that of P-TEFb (dCDK9 + dCyclin T). Chromatin immunoprecipitation (ChIP) experiments confirm that dCDK12 is present on the transcribed regions of active Drosophila genes. Compared with P-TEFb, dCDK12 amounts are lower at the 5' end and higher in the middle and at the 3' end of genes (both normalized to RNAPII). Appropriately, Drosophila dCDK12 purified from nuclear extracts manifests CTD kinase activity in vitro. Intriguingly, we find that cyclin K is associated with purified dCDK12, implicating it as the cyclin subunit of this CTD kinase. Most importantly, we demonstrate that RNAi knockdown of dCDK12 in S2 cells alters the phosphorylation state of the CTD, reducing its Ser2 phosphorylation levels. Similarly, in human HeLa cells, we show that hCDK13 purified from nuclear extracts displays CTD kinase activity in vitro, as anticipated. Also, we find that chimeric (yeast/human) versions of Ctk1 containing the kinase homology domains of hCDK12/13 (or hCDK9) are functional in yeast cells (and also in vitro); using this system, we show that a bur1(ts) mutant is rescued more efficiently by a hCDK9 chimera than by a hCDK13 chimera, suggesting the following orthology relationships: Bur1 ↔ CDK9 and Ctk1 ↔ CDK12/13. Finally, we show that siRNA knockdown of hCDK12 in HeLa cells results in alterations in the CTD phosphorylation state. Our findings demonstrate that metazoan CDK12 and CDK13 are CTD kinases, and that CDK12 is orthologous to yeast Ctk1.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Animales , Western Blotting , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Línea Celular , Mapeo Cromosómico , Ciclina T/genética , Ciclina T/metabolismo , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Células HeLa , Humanos , Microscopía Fluorescente , Mutación , Fosforilación , Proteínas Quinasas/genética , Interferencia de ARN , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
13.
Biochim Biophys Acta ; 1846(2): 638-54, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25450580

RESUMEN

mTOR (the mechanistic target of rapamycin) is an atypical serine/threonine kinase involved in regulating major cellular functions including growth and proliferation. Deregulation of the mTOR signaling pathway is one of the most commonly observed pathological alterations in human cancers. To this end, oncogenic activation of the mTOR signaling pathway contributes to cancer cell growth, proliferation and survival, highlighting the potential for targeting the oncogenic mTOR pathway members as an effective anti-cancer strategy. In order to do so, a thorough understanding of the physiological roles of key mTOR signaling pathway components and upstream regulators would guide future targeted therapies. Thus, in this review, we summarize available genetic mouse models for mTORC1 and mTORC2 components, as well as characterized mTOR upstream regulators and downstream targets, and assign a potential oncogenic or tumor suppressive role for each evaluated molecule. Together, our work will not only facilitate the current understanding of mTOR biology and possible future research directions, but more importantly, provide a molecular basis for targeted therapies aiming at key oncogenic members along the mTOR signaling pathway.


Asunto(s)
Carcinogénesis , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Autofagia , Proteínas Portadoras/fisiología , Humanos , Ratones , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/fisiología
14.
Int J Mol Sci ; 16(11): 26880-913, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26569225

RESUMEN

Ionizing radiation (IR), such as X-rays and gamma (γ)-rays, mediates various forms of cancer cell death such as apoptosis, necrosis, autophagy, mitotic catastrophe, and senescence. Among them, apoptosis and mitotic catastrophe are the main mechanisms of IR action. DNA damage and genomic instability contribute to IR-induced cancer cell death. Although IR therapy may be curative in a number of cancer types, the resistance of cancer cells to radiation remains a major therapeutic problem. In this review, we describe the morphological and molecular aspects of various IR-induced types of cell death. We also discuss cytogenetic variations representative of IR-induced DNA damage and genomic instability. Most importantly, we focus on several pathways and their associated marker proteins responsible for cancer resistance and its therapeutic implications in terms of cancer cell death of various types and characteristics. Finally, we propose radiation-sensitization strategies, such as the modification of fractionation, inflammation, and hypoxia and the combined treatment, that can counteract the resistance of tumors to IR.


Asunto(s)
Aberraciones Cromosómicas , Terapia Combinada/métodos , Rayos gamma/uso terapéutico , Neoplasias/terapia , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Apoptosis/genética , Apoptosis/efectos de la radiación , Autofagia/genética , Autofagia/efectos de la radiación , Análisis Citogenético , Daño del ADN/efectos de la radiación , Inestabilidad Genómica , Humanos , Mitosis/efectos de la radiación , Necrosis/genética , Necrosis/patología , Neoplasias/genética , Neoplasias/patología , Terapia por Rayos X
15.
IEEE Trans Cybern ; PP2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38354075

RESUMEN

This article focuses on the Pareto optimal issues of nonlinear game systems with asymmetric input saturation under dynamic event-triggered mechanism (DETM). First, the safe control is guaranteed by transforming the system with safety constraints into the one without state constraints utilizing barrier function. The united cost function integrating nonquadratic utility function is constructed to provide the foundation to achieve the Pareto optimal solutions. Then, the adaptive dynamic programming method with concurrent learning is proposed to approximate the Pareto optimal strategies wherein both current and historical data are utilized. To further lessen the consumptions of computation/communication resources, the DETM is integrated into the adaptive algorithm framework which can avoid Zeno phenomena. All the signals of the closed-loop system are proved to be uniformly ultimately bounded. Finally, the simulation results are given to validate the effectiveness of the proposed method from several aspects.

16.
Nat Commun ; 15(1): 5292, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38906855

RESUMEN

Ewing sarcoma is a pediatric bone and soft tissue tumor treated with chemotherapy, radiation, and surgery. Despite intensive multimodality therapy, ~50% patients eventually relapse and die of the disease due to chemoresistance. Here, using phospho-profiling, we find Ewing sarcoma cells treated with chemotherapeutic agents activate TAM (TYRO3, AXL, MERTK) kinases to augment Akt and ERK signaling facilitating chemoresistance. Mechanistically, chemotherapy-induced JAK1-SQ phosphorylation releases JAK1 pseudokinase domain inhibition allowing for JAK1 activation. This alternative JAK1 activation mechanism leads to STAT6 nuclear translocation triggering transcription and secretion of the TAM kinase ligand GAS6 with autocrine/paracrine consequences. Importantly, pharmacological inhibition of either JAK1 by filgotinib or TAM kinases by UNC2025 sensitizes Ewing sarcoma to chemotherapy in vitro and in vivo. Excitingly, the TAM kinase inhibitor MRX-2843 currently in human clinical trials to treat AML and advanced solid tumors, enhances chemotherapy efficacy to further suppress Ewing sarcoma tumor growth in vivo. Our findings reveal an Ewing sarcoma chemoresistance mechanism with an immediate translational value.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Janus Quinasa 1 , Proteínas Tirosina Quinasas Receptoras , Sarcoma de Ewing , Transducción de Señal , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Sarcoma de Ewing/genética , Humanos , Janus Quinasa 1/metabolismo , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Línea Celular Tumoral , Animales , Transducción de Señal/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ratones , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Tirosina Quinasa del Receptor Axl , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa c-Mer/metabolismo , Tirosina Quinasa c-Mer/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Fosforilación/efectos de los fármacos , Femenino , Factor de Transcripción STAT6
17.
ACS Appl Mater Interfaces ; 16(1): 669-683, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38150676

RESUMEN

Conventional photoresponsive materials have low photon utilization due to irregular distribution of photoactive groups, which severely limits the related real applications. Metal-organic frameworks (MOFs) can modulate the regular arrangement of functional groups to improve the electron transport paths and enhance the photon utilization, which provides strong support for the development of photoactive materials with excellent performance. In this work, one effective strategy for constructing a photoactive MOF had been developed via the utilization of Cd2+ and pyrazinoquinoxaline tetracarboxylic acid. The structural advantages of the Cd-MOF, such as a porous structure, abundant subject-object interaction sites, and a stable framework, ensure the prerequisite for various applications, while the better synergistic effect of Cd3 clusters and the pyrazinoquinoxaline derivative ensures efficient electron transfer efficiency. Therefore, by virtue of these structural advantages, the Cd-MOF can achieve fluorescence quenching detection for a variety of substrates, such as Fe3+, Cr2O72-, MnO4-, nitrofuran antibiotics, and TNP explosives, while fluorescence enhancement detection can be achieved for halogen ions, Cs+, Pb2+, and NO2-. In addition, the Cd-MOF can be used as a photocatalyst to successfully achieve the photocatalytic conversion of benzylamine to N-benzylbenzimidate under mild conditions. Thus, the Cd-MOF as a whole shows the possibility of application as a diverse fluorescence detection and photocatalyst and also illustrates the feasibility of preparing high-performance photoactive materials using the pyrazinoquinoxaline derivative.

18.
Biochim Biophys Acta ; 1825(1): 11-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21963805

RESUMEN

Prostate cancer is the most frequently diagnosed tumor in men and the second most common cause of cancer-related death for males in the United States. It has been shown that multiple signaling pathways are involved in the pathogenesis of prostate cancer, such as androgen receptor (AR), Akt, Wnt, Hedgehog (Hh) and Notch. Recently, burgeoning amounts of evidence have implicated that the F-box protein Skp2 (S-phase kinase associated protein 2), a well-characterized oncoprotein, also plays a critical role in the development and progression of prostate cancer. Therefore, this review discusses the recent literature regarding the function and regulation of Skp2 in the pathogenesis of prostate cancer. Furthermore, we highlight that Skp2 may represent an attractive therapeutic target, thus warrants further development of agents to target Skp2, which could have significant therapeutic impact on prostate cancer.


Asunto(s)
Neoplasias de la Próstata/terapia , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Terapia Molecular Dirigida , Neoplasias de la Próstata/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/fisiología , Transducción de Señal
19.
Bioessays ; 33(11): 851-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22006825

RESUMEN

Chemotherapy is an important therapeutic approach for cancer treatment. However, drug resistance is an obstacle that often impairs the successful use of chemotherapies. Therefore, overcoming drug resistance would lead to better therapeutic outcomes for cancer patients. Recently, studies by our own and other groups have demonstrated that there is an intimate correlation between the loss of the F-box and WD repeat domain-containing 7 (FBW7) tumor suppressor and the incurring drug resistance. While loss of FBW7 sensitizes cancer cells to certain drugs, FBW7-/- cells are more resistant to other types of chemotherapies. FBW7 exerts its tumor suppressor function by promoting the degradation of various oncoproteins that regulate many cellular processes, including cell cycle progression, cellular metabolism, differentiation, and apoptosis. Since loss of the FBW7 tumor suppressor is linked to drug resistance, FBW7 may represent a novel therapeutic target to increase drug sensitivity of cancer cells to conventional chemotherapeutics. This paper thus focuses on the new functional aspects of FBW7 in drug resistance.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Resistencia a Antineoplásicos , Proteínas F-Box/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Ubiquitina-Proteína Ligasas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Apoptosis , Bencenosulfonatos/farmacología , Compuestos de Bifenilo/farmacología , Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Sistema de Señalización de MAP Quinasas , MicroARNs/genética , MicroARNs/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Niacinamida/análogos & derivados , Nitrofenoles/farmacología , Paclitaxel/farmacología , Compuestos de Fenilurea , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Sorafenib , Sulfonamidas/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Vincristina/farmacología
20.
J Exp Med ; 220(10)2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37382881

RESUMEN

Somatic mutations occurring on key enzymes are extensively studied and targeted therapies are developed with clinical promises. However, context-dependent enzyme function through distinct substrates complicated targeting a given enzyme. Here, we develop an algorithm to elucidate a new class of somatic mutations occurring on enzyme-recognizing motifs that cancer may hijack to facilitate tumorigenesis. We validate BUD13-R156C and -R230Q mutations evading RSK3-mediated phosphorylation with enhanced oncogenicity in promoting colon cancer growth. Further mechanistic studies reveal BUD13 as an endogenous Fbw7 inhibitor that stabilizes Fbw7 oncogenic substrates, while cancerous BUD13-R156C or -R230Q interferes with Fbw7Cul1 complex formation. We also find this BUD13 regulation plays a critical role in responding to mTOR inhibition, which can be used to guide therapy selections. We hope our studies reveal the landscape of enzyme-recognizing motif mutations with a publicly available resource and provide novel insights for somatic mutations cancer hijacks to promote tumorigenesis with the potential for patient stratification and cancer treatment.


Asunto(s)
Neoplasias del Colon , Mutación con Ganancia de Función , Humanos , Carcinogénesis/genética , Transformación Celular Neoplásica , Mutación/genética , Proteínas de Unión al ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA