Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Genes Dev ; 35(1-2): 117-132, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33334825

RESUMO

The p53 tumor suppressor protein is a potent activator of proliferative arrest and cell death. In normal cells, this pathway is restrained by p53 protein degradation mediated by the E3-ubiquitin ligase activity of MDM2. Oncogenic stress releases p53 from MDM2 control, so activating the p53 response. However, many tumors that retain wild-type p53 inappropriately maintain the MDM2-p53 regulatory loop in order to continuously suppress p53 activity. We have shown previously that single point mutations in the human MDM2 RING finger domain prevent the interaction of MDM2 with the E2/ubiquitin complex, resulting in the loss of MDM2's E3 activity without preventing p53 binding. Here, we show that an analogous mouse MDM2 mutant (MDM2 I438K) restrains p53 sufficiently for normal growth but exhibits an enhanced stress response in vitro. In vivo, constitutive expression of MDM2 I438K leads to embryonic lethality that is rescued by p53 deletion, suggesting MDM2 I438K is not able to adequately control p53 function through development. However, the switch to I438K expression is tolerated in adult mice, sparing normal cells but allowing for an enhanced p53 response to DNA damage. Viewed as a proof of principle model for therapeutic development, our findings support an approach that would inhibit MDM2 E3 activity without preventing MDM2/p53 binding as a promising avenue for development of compounds to activate p53 in tumors with reduced on-target toxicities.


Assuntos
Desenvolvimento Embrionário/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Proliferação de Células/genética , Células Cultivadas , Embrião de Mamíferos/enzimologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Masculino , Camundongos , Mutação , Tamoxifeno/farmacologia
2.
Cell ; 149(6): 1183-5, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22682240

RESUMO

p53 is a key tumor suppressor protein that has numerous functions. Its primary mode of action has generally been ascribed to the induction of cell-cycle arrest, apoptosis, or senescence upon stress. Li et al. challenge this dogma with evidence that all three of these programs are dispensable for p53's tumor suppressive role.

4.
J Biol Chem ; 289(50): 34862-70, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25336640

RESUMO

Ubiquitin-specific peptidase 42 (USP42) is a deubiquitylating enzyme that can target p53 and contribute to the stabilization of p53 in response to stress. We now show that USP42 can also regulate transcription independently of p53. USP42 co-localized with RNA polymerase II (RNA Pol II) in nuclear foci, bound to histone H2B, and deubiquitylated H2B. Depletion of USP42 increased H2B ubiquitylation at a model promoter and decreased both basal and induced transcription from a number of promoters. These results are consistent with a role for USP42 in regulating transcription by deubiquitylating histones.


Assuntos
Regulação da Expressão Gênica , Histonas/metabolismo , Tioléster Hidrolases/metabolismo , Transcrição Gênica , Ubiquitinação , Transporte Ativo do Núcleo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Técnicas de Silenciamento de Genes , Humanos , Regiões Promotoras Genéticas/genética , Tioléster Hidrolases/deficiência , Tioléster Hidrolases/genética
5.
Biochim Biophys Acta ; 1843(1): 137-49, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23742843

RESUMO

The p53 tumor suppressor protein is involved in regulating a wide variety of stress responses, from senescence and apoptosis to more recently discovered roles in allowing adaptation to metabolic and oxidative stress. After 34years of research, significant progress has been made in unraveling the complexity of the p53 network, and it is clear that the regulation of p53 protein stability is critical in the control of p53 activity. This article focuses on our current understanding of how the level and activity of p53 is controlled by this seemingly simple mechanism. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.


Assuntos
Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Animais , Humanos , Processamento de Proteína Pós-Traducional/fisiologia , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Ativação Transcricional
6.
EMBO J ; 30(24): 4921-30, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22085928

RESUMO

The p53 tumour suppressor protein is a transcription factor that prevents oncogenic progression by activating the expression of apoptosis and cell-cycle arrest genes in stressed cells. The stability of p53 is tightly regulated by ubiquitin-dependent degradation, driven mainly by the ubiquitin ligase MDM2. In this study, we have identified USP42 as a DUB that interacts with and deubiquitinates p53. USP42 forms a direct complex with p53 and controls level of ubiquitination during the early phase of the response to a range of stress signals. Although we do not find a clear role for USP42 in controlling either the basal or fully activated levels of p53, the function of USP42 is required to allow the rapid activation of p53-dependent transcription and a p53-dependent cell-cycle arrest in response to stress. These functions of USP42 are likely to contribute to the repair and recovery of cells from mild or transient damage.


Assuntos
Tioléster Hidrolases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Humanos , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Interferente Pequeno/genética , Estresse Fisiológico , Tioléster Hidrolases/genética , Proteína Supressora de Tumor p53/genética
7.
Carcinogenesis ; 33(4): 791-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301280

RESUMO

p53 is a tumor suppressor that responds to a variety of stresses such as oncogenes and DNA damage by activating its transcriptional targets to allow repair or elimination of damaged cells. In the absence of stress signals, p53 needs to be kept in check and this is achieved by the E3 ligase MDM2. For tumors that retain wild-type p53, therapeutic strategies aimed at removing the inhibitory activity of MDM2 on p53 are under development and to date have focused on drugs that prevent the binding of p53 to MDM2. Here, we report the analysis of a group of synthetic analogs derived from 5-deazaflavin compounds previously identified in a screen as inhibitors of MDM2 autoubiquitination. Using measurement of surface plasmon resonance, we demonstrated that active 5-deazaflavin analogs bind to the MDM2 RING, whereas inactive compounds show no binding. In cellular assays, these active MDM2 RING binding compounds inhibited the ubiquitination of p53, stabilized p53, led to increased expression of p53 targets and caused corresponding cell cycle effects. Deazaflavin analogs therefore function to activate p53 through a novel mechanism, by inhibiting the E3 ligase activity of MDM2 in a manner that involves binding to the MDM2 RING.


Assuntos
Flavinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Flavinas/metabolismo , Citometria de Fluxo , Humanos , Ligação Proteica , Ressonância de Plasmônio de Superfície , Ubiquitinação
8.
Sci Signal ; 15(720): eabd9099, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35133863

RESUMO

Genetically encoded probes are widely used to visualize cellular processes in vitro and in vivo. Although effective in cultured cells, fluorescent protein tags and reporters are suboptimal in vivo because of poor tissue penetration and high background signal. Luciferase reporters offer improved signal-to-noise ratios but require injections of luciferin that can lead to variable responses and that limit the number and timing of data points that can be gathered. Such issues in studying the critical transcription factor p53 have limited insight on its activity in vivo during development and tissue injury responses. Here, by linking the expression of the near-infrared fluorescent protein iRFP713 to a synthetic p53-responsive promoter, we generated a knock-in reporter mouse that enabled noninvasive, longitudinal analysis of p53 activity in vivo in response to various stimuli. In the developing embryo, this model revealed the timing and localization of p53 activation. In adult mice, the model monitored p53 activation in response to irradiation and paracetamol- or CCl4-induced liver regeneration. After irradiation, we observed potent and sustained activation of p53 in the liver, which limited the production of reactive oxygen species (ROS) and promoted DNA damage resolution. We propose that this new reporter may be used to further advance our understanding of various physiological and pathophysiological p53 responses.


Assuntos
Regeneração Hepática , Proteína Supressora de Tumor p53 , Animais , Dano ao DNA , Genes Reporter , Regeneração Hepática/genética , Camundongos , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Nat Commun ; 12(1): 366, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446657

RESUMO

Many tumour cells show dependence on exogenous serine and dietary serine and glycine starvation can inhibit the growth of these cancers and extend survival in mice. However, numerous mechanisms promote resistance to this therapeutic approach, including enhanced expression of the de novo serine synthesis pathway (SSP) enzymes or activation of oncogenes that drive enhanced serine synthesis. Here we show that inhibition of PHGDH, the first step in the SSP, cooperates with serine and glycine depletion to inhibit one-carbon metabolism and cancer growth. In vitro, inhibition of PHGDH combined with serine starvation leads to a defect in global protein synthesis, which blocks the activation of an ATF-4 response and more broadly impacts the protective stress response to amino acid depletion. In vivo, the combination of diet and inhibitor shows therapeutic efficacy against tumours that are resistant to diet or drug alone, with evidence of reduced one-carbon availability. However, the defect in ATF4-response seen in vitro following complete depletion of available serine is not seen in mice, where dietary serine and glycine depletion and treatment with the PHGDH inhibitor lower but do not eliminate serine. Our results indicate that inhibition of PHGDH will augment the therapeutic efficacy of a serine depleted diet.


Assuntos
Glicina/metabolismo , Neoplasias/dietoterapia , Serina/biossíntese , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Glicina/análise , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/enzimologia , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Fosfoglicerato Desidrogenase/metabolismo , Serina/análise
10.
Nat Commun ; 11(1): 2094, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32350255

RESUMO

Phosphorylation of MDM2 by ATM upon DNA damage is an important mechanism for deregulating MDM2, thereby leading to p53 activation. ATM phosphorylates multiple residues near the RING domain of MDM2, but the underlying molecular basis for deregulation remains elusive. Here we show that Ser429 phosphorylation selectively enhances the ubiquitin ligase activity of MDM2 homodimer but not MDM2-MDMX heterodimer. A crystal structure of phospho-Ser429 (pS429)-MDM2 bound to E2-ubiquitin reveals a unique 310-helical feature present in MDM2 homodimer that allows pS429 to stabilize the closed E2-ubiquitin conformation and thereby enhancing ubiquitin transfer. In cells Ser429 phosphorylation increases MDM2 autoubiquitination and degradation upon DNA damage, whereas S429A substitution protects MDM2 from auto-degradation. Our results demonstrate that Ser429 phosphorylation serves as a switch to boost the activity of MDM2 homodimer and promote its self-destruction to enable rapid p53 stabilization and resolve a long-standing controversy surrounding MDM2 auto-degradation in response to DNA damage.


Assuntos
Dano ao DNA , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Domínios RING Finger , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Modelos Moleculares , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Multimerização Proteica , Relação Estrutura-Atividade , Ubiquitina/metabolismo
11.
Cell Rep ; 30(2): 481-496.e6, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31940491

RESUMO

Loss of p53 function contributes to the development of many cancers. While cell-autonomous consequences of p53 mutation have been studied extensively, the role of p53 in regulating the anti-tumor immune response is still poorly understood. Here, we show that loss of p53 in cancer cells modulates the tumor-immune landscape to circumvent immune destruction. Deletion of p53 promotes the recruitment and instruction of suppressive myeloid CD11b+ cells, in part through increased expression of CXCR3/CCR2-associated chemokines and macrophage colony-stimulating factor (M-CSF), and attenuates the CD4+ T helper 1 (Th1) and CD8+ T cell responses in vivo. p53-null tumors also show an accumulation of suppressive regulatory T (Treg) cells. Finally, we show that two key drivers of tumorigenesis, activation of KRAS and deletion of p53, cooperate to promote immune tolerance.


Assuntos
Células Mieloides/metabolismo , Linfócitos T Reguladores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Humanos , Camundongos
12.
Cancer Metab ; 6: 18, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524726

RESUMO

BACKGROUND: In response to oncogenic stress, the tumour suppressor protein p53 can induce the elimination of cells through induction of cell death or senescence, helping to restrain malignant progression. Conversely, under nutrient stress, p53 can protect cells by supporting metabolic adaptation. Many cancers express mutant p53 proteins that have lost the cell-elimination properties of wild-type p53. However, a previous report showed that a tumour-derived mutant can retain the ability to support cells under glutamine starvation. RESULTS: We show that a commonly occurring p53 mutant, R248W, retains wild-type ability to support survival under serine starvation. R248W, but not R175H, can engage p21 and MDM2, which both function to limit oxidative stress and facilitate the switch to de novo serine synthesis. In vivo, the growth of R248W-expressing tumours is resistant to dietary depletion of serine and glycine, correlating with an increased capacity to limit ROS compared to tumours expressing R175H. Human cancers expressing this p53 mutant show a worse outcome. CONCLUSION: Our work shows that mutant p53s can selectively retain wild-type p53 functions that allow adaptation to serine starvation through the activation of antioxidant defence pathways. Tumours containing this p53 mutation are resistant to serine-limited conditions and less responsive to therapy.

13.
Cell Metab ; 28(5): 721-736.e6, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30122553

RESUMO

Numerous mechanisms to support cells under conditions of transient nutrient starvation have been described. Several functions of the tumor-suppressor protein p53 can contribute to the adaptation of cells to metabolic stress and help cancer cell survival under nutrient-limiting conditions. We show here that p53 promotes the expression of SLC1A3, an aspartate/glutamate transporter that allows the utilization of aspartate to support cells in the absence of extracellular glutamine. Under glutamine deprivation, SLC1A3 expression maintains electron transport chain and tricarboxylic acid cycle activity, promoting de novo glutamate, glutamine, and nucleotide synthesis to rescue cell viability. Tumor cells with high levels of SLC1A3 expression are resistant to glutamine starvation, and SLC1A3 depletion retards the growth of these cells in vitro and in vivo, suggesting a therapeutic potential for SLC1A3 inhibition.


Assuntos
Transportador 1 de Aminoácido Excitatório/metabolismo , Glutamina/metabolismo , Neoplasias/metabolismo , Inanição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adaptação Fisiológica , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclo do Ácido Cítrico , Feminino , Humanos , Camundongos Endogâmicos BALB C
14.
Nat Struct Mol Biol ; 24(7): 578-587, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28553961

RESUMO

MDM2-MDMX complexes bind the p53 tumor-suppressor protein, inhibiting p53's transcriptional activity and targeting p53 for proteasomal degradation. Inhibitors that disrupt binding between p53 and MDM2 efficiently activate a p53 response, but their use in the treatment of cancers that retain wild-type p53 may be limited by on-target toxicities due to p53 activation in normal tissue. Guided by a novel crystal structure of the MDM2-MDMX-E2(UbcH5B)-ubiquitin complex, we designed MDM2 mutants that prevent E2-ubiquitin binding without altering the RING-domain structure. These mutants lack MDM2's E3 activity but retain the ability to limit p53's transcriptional activity and allow cell proliferation. Cells expressing these mutants respond more quickly to cellular stress than cells expressing wild-type MDM2, but basal p53 control is maintained. Targeting the MDM2 E3-ligase activity could therefore widen the therapeutic window of p53 activation in tumors.


Assuntos
Proteólise , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular , Cristalografia por Raios X , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/genética , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo
15.
Sci Rep ; 7(1): 1837, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28500323

RESUMO

While the use of bioluminescent proteins for molecular imaging is a powerful technology to further our understanding of complex processes, fluorescent labeling with visible light fluorescent proteins such as GFP and RFP suffers from poor tissue penetration and high background autofluorescence. To overcome these limitations, we generated an inducible knock-in mouse model of iRFP713. This model was used to assess Cre activity in a Rosa Cre-ER background and quantify Cre activity upon different tamoxifen treatments in several organs. We also show that iRFP can be readily detected in 3D organoid cultures, FACS analysis and in vivo tumour models. Taken together we demonstrate that iRFP713 is a progressive step in in vivo imaging and analysis that widens the optical imaging window to the near-infrared spectrum, thereby allowing deeper tissue penetration, quicker image acquisition without the need to inject substrates and a better signal to background ratio in genetically engineered mouse models (GEMMs).


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Proteínas Luminescentes/genética , Neoplasias/genética , Neoplasias/metabolismo , Recombinases/metabolismo , Animais , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Expressão Gênica , Genes Reporter , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Imagem Molecular/métodos , Neoplasias/patologia , Imagem Óptica/métodos
16.
Nat Cell Biol ; 17(12): 1556-68, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26595383

RESUMO

L-Glutamine (Gln) functions physiologically to balance the carbon and nitrogen requirements of tissues. It has been proposed that in cancer cells undergoing aerobic glycolysis, accelerated anabolism is sustained by Gln-derived carbons, which replenish the tricarboxylic acid (TCA) cycle (anaplerosis). However, it is shown here that in glioblastoma (GBM) cells, almost half of the Gln-derived glutamate (Glu) is secreted and does not enter the TCA cycle, and that inhibiting glutaminolysis does not affect cell proliferation. Moreover, Gln-starved cells are not rescued by TCA cycle replenishment. Instead, the conversion of Glu to Gln by glutamine synthetase (GS; cataplerosis) confers Gln prototrophy, and fuels de novo purine biosynthesis. In both orthotopic GBM models and in patients, (13)C-glucose tracing showed that GS produces Gln from TCA-cycle-derived carbons. Finally, the Gln required for the growth of GBM tumours is contributed only marginally by the circulation, and is mainly either autonomously synthesized by GS-positive glioma cells, or supplied by astrocytes.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células , Glioblastoma/metabolismo , Glutamato-Amônia Ligase/metabolismo , Glutamina/metabolismo , Nucleotídeos/biossíntese , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Células Cultivadas , Ciclo do Ácido Cítrico , Técnicas de Cocultura , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glutamato-Amônia Ligase/genética , Ácido Glutâmico/metabolismo , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Células-Tronco Neoplásicas/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
17.
Cell Cycle ; 13(2): 220-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24200967

RESUMO

GFP and luciferase are used extensively as markers both in vitro and in vivo although both have limitations. The utility of GFP fluorescence is restricted by high background signal and poor tissue penetrance. Luciferase throughput is limited in vitro by the requirement for cell lysis, while in vivo, luciferase readout is complicated by the need for substrate injection and the dependence on endogenous ATP. Here we show that near-infrared fluorescent protein in combination with widely available near-infrared scanners overcomes these obstacles and allows for the accurate determination of cell number in vitro and tumor growth in vivo in a high-throughput manner and at negligible per-well costs. This system represents a significant advance in tracking cell proliferation in tissue culture as well as in animals, with widespread applications in cell biology.


Assuntos
Biomarcadores Tumorais/metabolismo , Proliferação de Células , Proteínas Luminescentes/metabolismo , Neoplasias Experimentais/patologia , Células 3T3 , Animais , Contagem de Células , Linhagem Celular Transformada , Linhagem Celular Tumoral , Feminino , Fatores de Transcrição Forkhead/genética , Xenoenxertos , Ensaios de Triagem em Larga Escala , Humanos , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Espectroscopia de Luz Próxima ao Infravermelho
18.
PLoS One ; 9(6): e98399, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24887316

RESUMO

Anchorage independent growth is one of the hallmarks of oncogenic transformation. Here we show that infrared fluorescent protein (iRFP) based assays allow accurate and unbiased determination of colony formation and anchorage independent growth over time. This protocol is particularly compatible with high throughput systems, in contrast to traditional methods which are often labor-intensive, subjective to bias and do not allow further analysis using the same cells. Transformation in a single layer soft agar assay could be documented as early as 2 to 3 days in a 96 well format, which can be easily combined with standard transfection, infection and compound screening setups to allow for high throughput screening to identify therapeutic targets.


Assuntos
Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica , Proteínas Luminescentes/metabolismo , Células 3T3 , Animais , Camundongos
19.
Autophagy ; 10(12): 2269-78, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25629932

RESUMO

During macroautophagy, conjugation of ATG12 to ATG5 is essential for LC3 lipidation and autophagosome formation. Additionally, ATG12 has ATG5-independent functions in diverse processes including mitochondrial fusion and mitochondrial-dependent apoptosis. In this study, we investigated the regulation of free ATG12. In stark contrast to the stable ATG12-ATG5 conjugate, we find that free ATG12 is highly unstable and rapidly degraded in a proteasome-dependent manner. Surprisingly, ATG12, itself a ubiquitin-like protein, is directly ubiquitinated and this promotes its proteasomal degradation. As a functional consequence of its turnover, accumulation of free ATG12 contributes to proteasome inhibitor-mediated apoptosis, a finding that may be clinically important given the use of proteasome inhibitors as anticancer agents. Collectively, our results reveal a novel interconnection between autophagy, proteasome activity, and cell death mediated by the ubiquitin-like properties of ATG12.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Mitocôndrias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Animais , Proteína 12 Relacionada à Autofagia , Morte Celular/fisiologia , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Enzimas de Conjugação de Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa