Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 535
Filtrar
1.
Drug Des Devel Ther ; 15: 5061-5074, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34938069

RESUMO

INTRODUCTION: Sunitinib is the first-line targeted drug for the treatment of advanced renal cell carcinoma (RCC), but its therapeutic potential is limited by premature drug resistance. In an attempt to overcome this limitation, a sunitinib-resistant cell-derived xenograft (CDX) model of clear cell renal cell carcinoma (ccRCC) in vivo was constructed in this study. The molecular mechanism of drug resistance was analyzed using sequencing and bioinformatics tools. METHODS: First, mice were injected subcutaneously with tumor cells 786-O to create tumors and were simultaneously treated with sunitinib. After three consecutive passages, a drug-resistant xenograft model was obtained. Then, key pathways and genes were identified via second-generation sequencing of the tissue and the examination of differentially expressed genes (DEGs) with bioinformatics tools. RESULTS: Analysis of sequencing data revealed that 646 DEGs were upregulated and 465 were downregulated in the drug-resistant tissues when compared with the sensitive tissues. GO showed that the DEGs were significantly enriched in angiogenesis, cell hypoxia response, and apoptosis. KEGG analysis demonstrated that the main pathways were PI3K-Akt, HIF-1, NF-kappa B, and MAPK. Modular analysis of the PPI network indicated that the GO and KEGG analyses of module 1 with the highest ranking were mainly related to ubiquitinase activity. Similarly, the GO and KEGG analyses of the top 10 hub genes were also chiefly linked to ubiquitinase activity. Then, comprehensive expression analysis of the hub genes, and finally, the genes BTRC and TRIM32 were identified, which were consistent in all observations. CONCLUSION: In this study, through the construction of in vitro models and bioinformatics analysis, the important pathways and key genes related to ccRCC sunitinib resistance were discovered. Among them, ubiquitinase may play an important role in drug resistance and may be a potential therapeutic target and biomarker.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Renais/tratamento farmacológico , Sunitinibe/farmacologia , Animais , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Biologia Computacional , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Neoplasias Renais/genética , Camundongos , Ubiquitina/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Sci Rep ; 11(1): 23056, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-34845242

RESUMO

Cardiogenesis requires the orchestrated spatiotemporal tuning of BMP signalling upon the balance between induction and counter-acting suppression of the differentiation of the cardiac tissue. SMADs are key intracellular transducers and the selective degradation of SMADs by the ubiquitin-proteasome system is pivotal in the spatiotemporal tuning of BMP signalling. However, among three SMADs for BMP signalling, SMAD1/5/9, only the specific E3 ligase of SMAD9 remains poorly investigated. Here, we report for the first time that SMAD9, but not the other SMADs, is ubiquitylated by the E3 ligase ASB2 and targeted for proteasomal degradation. ASB2, as well as Smad9, is conserved among vertebrates. ASB2 expression was specific to the cardiac region from the very early stage of cardiac differentiation in embryogenesis of mouse. Knockdown of Asb2 in zebrafish resulted in a thinned ventricular wall and dilated ventricle, which were rescued by simultaneous knockdown of Smad9. Abundant Smad9 protein leads to dysregulated cardiac differentiation through a mechanism involving Tbx2, and the BMP signal conducted by Smad9 was downregulated under quantitative suppression of Smad9 by Asb2. Our findings demonstrate that ASB2 is the E3 ligase of SMAD9 and plays a pivotal role in cardiogenesis through regulating BMP signalling.


Assuntos
Coração , Proteína Smad8 , Proteínas Supressoras da Sinalização de Citocina , Animais , Humanos , Camundongos , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Células HEK293 , Análise de Sequência com Séries de Oligonucleotídeos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Proteína Smad8/biossíntese , Proteína Smad8/fisiologia , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Ubiquitina/química , Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
3.
Infect Immun ; 89(11): e0020221, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34338548

RESUMO

Gamma interferon (IFN-γ)-induced immunity-related GTPases (IRGs) confer cell-autonomous immunity to the intracellular protozoan pathogen Toxoplasma gondii. Effector IRGs are loaded onto the Toxoplasma-containing parasitophorous vacuole (PV), where they recruit ubiquitin ligases, ubiquitin-binding proteins, and IFN-γ-inducible guanylate-binding proteins (Gbps), prompting PV lysis and parasite destruction. Host cells lacking the regulatory IRGs Irgm1 and Irgm3 fail to load effector IRGs, ubiquitin, and Gbps onto the PV and are consequently defective for cell-autonomous immunity to Toxoplasma. However, the role of the third regulatory IRG, Irgm2, in cell-autonomous immunity to Toxoplasma has remained unexplored. Here, we report that Irgm2 unexpectedly plays a limited role in the targeting of effector IRGs, ubiquitin, and Gbps to the Toxoplasma PV. Instead, Irgm2 is instrumental in the decoration of PVs with γ-aminobutyric acid receptor-associated protein-like 2 (GabarapL2). Cells lacking Irgm2 are as defective for cell-autonomous host defense to Toxoplasma as pan-Irgm-/- cells lacking all three Irgm proteins, and Irgm2-/- mice succumb to Toxoplasma infections as readily as pan-Irgm-/- mice. These findings demonstrate that, relative to Irgm1 and Irgm3, Irgm2 plays a distinct but critically important role in host resistance to Toxoplasma.


Assuntos
GTP Fosfo-Hidrolases/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Toxoplasmose/imunologia , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/fisiologia , Ubiquitina/fisiologia , Vacúolos/fisiologia
4.
Cell Microbiol ; 23(2): e13276, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33037857

RESUMO

Viruses confiscate cellular components of the ubiquitin-proteasome system (UPS) to facilitate many aspects of the infectious cycle. The 26S proteasome is an ATP-dependent, multisubunit proteolytic machine present in all eukaryotic cells. The proteasome executes the controlled degradation of functional proteins, as well as the hydrolysis of aberrantly folded polypeptides. There is growing evidence for the role of the UPS in viral entry. The UPS assists in several steps of the initiation of infection, including endosomal escape of the entering virion, intracellular transport of incoming nucleocapsids and uncoating of the viral genome. Inhibitors of proteasome activity, including MG132, epoxomicin, lactacystin and bortezomib have been integral to developments in this area. Here, we review the mechanistic details of UPS involvement in the entry process of viruses from a multitude of families. The possibility of proteasome inhibitors as therapeutic antiviral agents is highlighted.


Assuntos
Interações entre Hospedeiro e Microrganismos , Complexo de Endopeptidases do Proteassoma/fisiologia , Inibidores de Proteassoma/farmacologia , Ubiquitina/fisiologia , Internalização do Vírus , Fenômenos Fisiológicos Virais , Vírus/efeitos dos fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Antivirais/farmacologia , Bortezomib/farmacologia , Humanos , Leupeptinas/farmacologia , Nucleocapsídeo/metabolismo , Oligopeptídeos/farmacologia , Proteólise , Vírion/metabolismo
5.
Nucleic Acids Res ; 49(2): 902-915, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33348378

RESUMO

Repair of covalent DNA-protein crosslinks (DPCs) by the metalloprotease SPRTN prevents genome instability, premature aging and carcinogenesis. SPRTN is specifically activated by DNA structures containing single- and double-stranded features, but degrades the protein components of DPCs promiscuously and independent of amino acid sequence. This lack of specificity is useful to target diverse protein adducts, however, it requires tight control in return, in order to prohibit uncontrolled proteolysis of chromatin proteins. Here, we discover the components and principles of a ubiquitin switch, which negatively regulates SPRTN. We demonstrate that monoubiquitylation is induced in an E3 ligase-independent manner and, in contrast to previous assumptions, does not control chromatin access of the enzyme. Data obtained in cells and in vitro reveal that monoubiquitylation induces inactivation of the enzyme by triggering autocatalytic cleavage in trans while also priming SPRTN for proteasomal degradation in cis. Finally, we show that the deubiquitylating enzyme USP7 antagonizes this negative control of SPRTN in the presence of DPCs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Processamento de Proteína Pós-Traducional , Ubiquitina/fisiologia , Ubiquitinação , Catálise , Linhagem Celular , Cromatina/metabolismo , Adutos de DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/química , Enzimas Desubiquitinantes/metabolismo , Técnicas de Inativação de Genes , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Peptidase 7 Específica de Ubiquitina/fisiologia
6.
Elife ; 92020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32573437

RESUMO

RNF213 is the major susceptibility factor for Moyamoya disease, a progressive cerebrovascular disorder that often leads to brain stroke in adults and children. Characterization of disease-associated mutations has been complicated by the enormous size of RNF213. Here, we present the cryo-EM structure of mouse RNF213. The structure reveals the intricate fold of the 584 kDa protein, comprising an N-terminal stalk, a dynein-like core with six ATPase units, and a multidomain E3 module. Collaboration with UbcH7, a cysteine-reactive E2, points to an unexplored ubiquitin-transfer mechanism that proceeds in a RING-independent manner. Moreover, we show that pathologic MMD mutations cluster in the composite E3 domain, likely interfering with substrate ubiquitination. In conclusion, the structure of RNF213 uncovers a distinct type of an E3 enzyme, highlighting the growing mechanistic diversity in ubiquitination cascades. Our results also provide the molecular framework for investigating the emerging role of RNF213 in lipid metabolism, hypoxia, and angiogenesis.


Moyamoya disease is a genetic disorder affecting both adults and children. It is characterized by narrowing of the blood vessels in the brain, which can lead to strokes. Moyamoya patients often have mutations in the gene for a protein called RNF213. This protein is linked to multiple processes in the body, including the development of blood vessels. Despite this, its role in Moyamoya disease is still something of a mystery. RNF213 is known to fall into two protein 'classes'. First, it is an E3 enzyme. This type of protein tags unwanted or defective proteins for disposal by the cell. Second, it is a motor protein. Motor proteins contain tiny molecular 'engines', called ATPases, that normally convert chemical energy to movement. No other human protein combines these two activities, making RNF213 unique. RNF213 is also an extremely large protein, which means it is difficult to manipulate in the laboratory and thus hard to study. Scientists still need more detailed information on RNF213's structure and chemical activity before we can understand what the mutant protein might be doing in Moyamoya disease. Ahel et al. therefore set out to make the RNF213 protein and 'dissect' it in a test tube. Electron microscopy experiments using the mouse-version of RNF213 revealed that it consisted of a single, giant molecule, folded up to form three regions with distinct structures. These were a long 'arm' at one end, a ring-shaped part in the middle, containing the ATPase 'motor', and the E3 enzyme module at the other end. Further chemical analysis showed that RNF213's ATPase and E3 modules worked in unexpected ways. Although the ATPase did resemble another well-known motor protein, in RNF213 it did not generate movement but rather appeared to act like an intricate molecular 'switch'. The E3 module of RNF213 'tagged' other molecules as expected but did not contain an additional structure that all other known E3 enzymes need to work properly. This suggests that RNF213 represents a distinct class of E3 enzymes. Biochemical tests of the mutation most commonly found in Moyamoya patients revealed that it left RNF213's overall structure, ATPase motor and E3 module intact. That is, the disease-causing mutation appeared to hinder interactions with other partner proteins, rather than disrupting RNF213 itself. By providing the first detailed molecular description of the architecture of RNF213, Ahel et al. hope that these findings will help future investigations of both this giant protein's biological role in the cell and its contribution to Moyamoya disease.


Assuntos
Adenosina Trifosfatases/genética , Doença de Moyamoya/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina/fisiologia , Adenosina Trifosfatases/química , Animais , Camundongos , Doença de Moyamoya/patologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/química
7.
Ageing Res Rev ; 61: 101078, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32407951

RESUMO

The abnormal accumulation of neurotoxic proteins is the typical hallmark of various age-related neurodegenerative disorders (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis and Multiple sclerosis. The anomalous proteins, such as Aß, Tau in Alzheimer's disease and α-synuclein in Parkinson's disease, perturb the neuronal physiology and cellular homeostasis in the brain thereby affecting the millions of human lives across the globe. Here, ubiquitin proteasome system (UPS) plays a decisive role in clearing the toxic metabolites in cells, where any aberrancy is widely reported to exaggerate the neurodegenerative pathologies. In spite of well-advancement in the ubiquitination research, their molecular markers and mechanisms for target-specific protein ubiquitination and clearance remained elusive. Therefore, this review substantiates the role of UPS in the brain signaling and neuronal physiology with their mechanistic role in the NDD's specific pathogenic protein clearance. Moreover, current and future promising therapies are discussed to target UPS-mediated neurodegeneration for better public health.


Assuntos
Doenças Neurodegenerativas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/fisiologia , Doença de Alzheimer/metabolismo , Humanos , Transdução de Sinais , Ubiquitina/metabolismo , alfa-Sinucleína
9.
Neurobiol Aging ; 91: 160-166, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32280031

RESUMO

Brain aging is accompanied by an accumulation of damaged proteins, which results from deterioration of cellular quality control mechanisms and decreased protein degradation. The ubiquitin-proteasome system (UPS) is the primary proteolytic mechanism responsible for targeted degradation. Recent work has established a critical role of the UPS in memory and synaptic plasticity, but the role of the UPS in age-related cognitive decline remains poorly understood. Here, we measured markers of UPS function and related them to fear memory in rats. Our results show that age-related memory deficits are associated with reductions in phosphorylation of the Rpt6 proteasome regulatory subunit and corresponding increases in lysine-48 (K48)-linked ubiquitin tagging within the basolateral amygdala. Increases in K48 polyubiquitination were also observed in the medial prefrontal cortex and dorsal hippocampus. These data suggest that protein degradation is a critical component of age-related memory deficits. This extends our understanding of the relationship between the UPS, aging, and memory, which is an important step toward the prevention and treatment of deficits associated with normal cognitive aging and memory-related neurodegenerative diseases.


Assuntos
Tonsila do Cerebelo/metabolismo , Envelhecimento Cognitivo/psicologia , Condicionamento Clássico , Medo/fisiologia , Hipocampo/metabolismo , Transtornos da Memória/etiologia , Memória/fisiologia , Córtex Pré-Frontal/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteólise , Ubiquitina/fisiologia , Animais , Masculino , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos Endogâmicos F344 , Ubiquitina/metabolismo
10.
Sci Rep ; 10(1): 5689, 2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32231214

RESUMO

Drosophila melanogaster is a useful and highly tractable model organism for understanding the molecular mechanisms of human diseases. We previously characterized a new dUbqn knockdown model that induces learning-memory and locomotive deficits mediated by impaired proteostasis. Although proteinopathies are the main causes of neurodegenerative diseases, limited information is currently available on the relationship between proteostasis and neurodegenerative-related behavioral perturbations, such as locomotion, wakefulness, and sexual activities. Thus, the present study aimed to elucidate the mechanisms by which dUbqn depletion which is known to cause proteinopathies, affects neurodegenerative-related behavioral perturbations. Pan-neuronal dUbqn-depleted flies showed significantly reduced evening activity along with altered pre- and postsynaptic structural NMJ's proteins by attenuating signals of Bruchpilot puncta and GluRIIA clustering. In addition, the neurochemical profiles of GABA, glutamate, dopamine, and serotonin were disturbed and these changes also affected courtship behaviors in dUbqn-depleted flies. Collectively, these results extend our understanding on how dUbqn depletion affects neurochemical regulation to drive behavioral disturbances that are generally found in the early stage of neurodegenerative diseases. Moreover, the present study may contribute a novel finding to the design of new agents that prevent disease progression or even treat diseases related to neurodegeneration.


Assuntos
Drosophila melanogaster/metabolismo , Ubiquitina/deficiência , Animais , Corte , Dopamina/metabolismo , Dopamina/fisiologia , Drosophila melanogaster/fisiologia , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Ácido Glutâmico/metabolismo , Ácido Glutâmico/fisiologia , Larva , Masculino , Espectrometria de Massas , Atividade Motora/fisiologia , Sistema Nervoso/metabolismo , Sistema Nervoso/fisiopatologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Serotonina/metabolismo , Serotonina/fisiologia , Espectrometria de Massas em Tandem , Ubiquitina/metabolismo , Ubiquitina/fisiologia , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/fisiologia
11.
Biochem Soc Trans ; 48(2): 479-497, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32196552

RESUMO

Eukaryotic life depends upon the interplay between vast networks of signaling pathways composed of upwards of 109-1010 proteins per cell. The integrity and normal operation of the cell requires that these proteins act in a precise spatial and temporal manner. The ubiquitin system is absolutely central to this process and perturbation of its function contributes directly to the onset and progression of a wide variety of diseases, including cancer, metabolic syndromes, neurodegenerative diseases, autoimmunity, inflammatory disorders, infectious diseases, and muscle dystrophies. Whilst the individual components and the overall architecture of the ubiquitin system have been delineated in some detail, how ubiquitination might be successfully targeted, or harnessed, to develop novel therapeutic approaches to the treatment of disease, currently remains relatively poorly understood. In this review, we will provide an overview of the current status of selected small molecule ubiquitin system inhibitors. We will further discuss the unique challenges of targeting this ubiquitous and highly complex machinery, and explore and highlight potential ways in which these challenges might be met.


Assuntos
Ubiquitina/fisiologia , Ubiquitinação , Autoimunidade , Doenças Transmissíveis/metabolismo , Desenho de Fármacos , Descoberta de Drogas , Humanos , Inflamação/metabolismo , Síndrome Metabólica/metabolismo , Modelos Biológicos , Distrofias Musculares/metabolismo , Neoplasias/metabolismo , Doenças Neurodegenerativas/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Inibidores de Proteassoma/uso terapêutico , Processamento de Proteína Pós-Traducional , Transdução de Sinais
12.
Lancet Psychiatry ; 7(6): 528-537, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32061320

RESUMO

The ubiquitin-proteasome system is a master regulator of neural development and the maintenance of brain structure and function. It influences neurogenesis, synaptogenesis, and neurotransmission by determining the localisation, interaction, and turnover of scaffolding, presynaptic, and postsynaptic proteins. Moreover, ubiquitin-proteasome system signalling transduces epigenetic changes in neurons independently of protein degradation and, as such, dysfunction of components and substrates of this system has been linked to a broad range of brain conditions. Although links between ubiquitin-proteasome system dysfunction and neurodegenerative disorders have been known for some time, only recently have similar links emerged for neurodevelopmental disorders, such as schizophrenia. Here, we review the components of the ubiquitin-proteasome system that are reported to be dysregulated in schizophrenia, and discuss specific molecular changes to these components that might, in part, explain the complex causes of this mental disorder.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Esquizofrenia/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina/metabolismo , Animais , Humanos , Modelos Animais , Doenças Neurodegenerativas/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Terminações Pré-Sinápticas/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Esquizofrenia/tratamento farmacológico , Esquizofrenia/fisiopatologia , Transmissão Sináptica/fisiologia , Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/uso terapêutico
13.
Neurotox Res ; 37(1): 48-57, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31654383

RESUMO

Motor neuron diseases are neurodegenerative diseases that are characterized by degeneration of the upper and lower motor neurons in the central nervous system. Mutations in Dynactin 1 (DCTN1), a component in the Dynein/Dynactin motor complex, have been previously identified to cause motor neuron diseases and other neurodegenerative disorders. Recent studies showed that motor neuron disease-linked mutation, such as G59S mutation, could lead to dysfunction and protein aggregation of DCTN1. However, the cellular pathway involved in the clearance of DCTN1 aggregates is still not fully elucidated. In this study, we employed a culture cell model of DCTN1-linked neurodegeneration and explored the role of cellular protein control systems in the regulation of wild type and mutant DCTN1. We find that the ubiquitin-proteasome system, but not autophagy, is the primary protein degradation system for the turnover of both wild type and G59S DCTN1 under normal conditions. However, it turns out that autophagy can play a role in the clearance of protein aggregates of G59S DCTN1 when the proteasome activity is inhibited. Importantly, overexpression of TFEB, a master regulator of autophagy, promotes the autophagic clearance of G59S DCTN1 aggregates and ameliorates G59S DCTN1-induced cytotoxicity when the proteasomes are impaired. In conclusion, autophagy may play as a backup system to protect cells against the cytotoxicity induced by aggregate-prone DCTN1 when proteasomal function is damaged.


Assuntos
Autofagia/fisiologia , Complexo Dinactina/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteólise , Ubiquitina/fisiologia , Animais , Sobrevivência Celular , Células Cultivadas , Complexo Dinactina/genética , Humanos , Camundongos , Microtúbulos/metabolismo , Mutação , Plasmídeos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Transfecção
15.
Science ; 366(6467): 818-822, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31727826

RESUMO

To achieve homeostasis, cells evolved dynamic and self-regulating quality control processes to adapt to new environmental conditions and to prevent prolonged damage. We discuss the importance of two major quality control systems responsible for degradation of proteins and organelles in eukaryotic cells: the ubiquitin-proteasome system (UPS) and autophagy. The UPS and autophagy form an interconnected quality control network where decision-making is self-organized on the basis of biophysical parameters (binding affinities, local concentrations, and avidity) and compartmentalization (through membranes, liquid-liquid phase separation, or the formation of aggregates). We highlight cellular quality control factors that delineate their differential deployment toward macromolecular complexes, liquid-liquid phase-separated subcellular structures, or membrane-bound organelles. Finally, we emphasize the need for continuous promotion of quantitative and mechanistic research into the roles of the UPS and autophagy in human pathophysiology.


Assuntos
Autofagia , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteólise , Ubiquitina/fisiologia , Animais , Humanos , Macroautofagia , Organelas/metabolismo
16.
Cells ; 8(8)2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31394841

RESUMO

Ubiquitin is a highly conserved small eukaryotic protein. It is generated by proteolytic cleavage of precursor proteins in which it is fused either to itself, constituting a polyubiquitin precursor of head-to-tail monomers, or as a single N-terminal moiety to ribosomal proteins. Understanding the role of the ubiquitin fused to ribosomal proteins becomes relevant, as these proteins are practically invariably eS31 and eL40 in the different eukaryotes. Herein, we used the amenable yeast Saccharomyces cerevisiae to study whether ubiquitin facilitates the expression of the fused eL40 (Ubi1 and Ubi2 precursors) and eS31 (Ubi3 precursor) ribosomal proteins. We have analyzed the phenotypic effects of a genomic ubi1∆ub-HA ubi2∆ mutant, which expresses a ubiquitin-free HA-tagged eL40A protein as the sole source of cellular eL40. This mutant shows a severe slow-growth phenotype, which could be fully suppressed by increased dosage of the ubi1∆ub-HA allele, or partially by the replacement of ubiquitin by the ubiquitin-like Smt3 protein. While expression levels of eL40A-HA from ubi1∆ub-HA are low, eL40A is produced practically at normal levels from the Smt3-S-eL40A-HA precursor. Finally, we observed enhanced aggregation of eS31-HA when derived from a Ubi3∆ub-HA precursor and reduced aggregation of eL40A-HA when expressed from a Smt3-S-eL40A-HA precursor. We conclude that ubiquitin might serve as a cis-acting molecular chaperone that assists in the folding and synthesis of the fused eL40 and eS31 ribosomal proteins.


Assuntos
Proteínas Ribossômicas/metabolismo , Saccharomyces cerevisiae/metabolismo , Ubiquitina/fisiologia
17.
FEBS J ; 286(14): 2711-2725, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30963698

RESUMO

Rev1, a Y-family DNA polymerase, is involved in the tolerance of DNA damage by translesion DNA synthesis (TLS). Previous studies have shown that the C-terminal domain (CTD) and ubiquitin (Ub)-binding (UBM) domains of Rev1 play important roles in UV-damage tolerance, but how these domains contribute to the process remains unclear. In this study, we created Ub mutations in a proliferating cell nuclear antigen (PCNA)-Ub fusion that differentially affect its interaction with Rev1 and Polη and found that UV-damage tolerance depends on its interaction with Rev1 but not Polη. We also created Rev1-UBM mutations altering its interaction with a PCNA-Ub fusion and Rev1-CTD mutations affecting its interaction with Polη and the Rev7 subunit of Polζ. We thus demonstrated that elevated expression of Rev1 alone is sufficient to confer enhanced UV-damage tolerance and that this tolerance depends on its physical interaction with monoubiquitinated PCNA and Polζ but is independent of Polη. Collectively, these studies reveal central roles played by Rev1 in coordinating UV-damage response pathway choice in mammalian cells.


Assuntos
Dano ao DNA , Nucleotidiltransferases/fisiologia , Raios Ultravioleta , DNA Polimerase Dirigida por DNA/fisiologia , Células HCT116 , Humanos , Mutação , Nucleotidiltransferases/química , Nucleotidiltransferases/genética , Antígeno Nuclear de Célula em Proliferação/fisiologia , Ubiquitina/fisiologia
18.
Neurosci Lett ; 703: 68-78, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-30890471

RESUMO

Several lines of evidence have shown that defects in the endo-lysosomal autophagy degradation pathway and the ubiquitin-proteasome system play a role in Alzheimer's Disease (AD) pathogenesis and pathophysiology. Early pathological changes, such as marked enlargement of endosomal compartments, gradual accumulation of autophagic vacuoles (AVs) and lysosome dyshomeostasis, are well-recognized in AD. In addition to these pathological indicators, many genetic variants of key regulators in the endo-lysosomal autophagy networks and the ubiquitin-proteasome system have been found to be associated with AD. Furthermore, altered expression levels of key proteins in these pathways have been found in AD human brain tissues, primary cells and AD mouse models. In this review, we discuss potential disease mechanisms underlying the dysregulation of protein homeostasis governing systems. While the importance of two major protein degradation pathways in AD pathogenesis has been highlighted, targeted therapy at key components of these pathways has great potential in developing novel therapeutic interventions for AD. Future investigations are needed to define molecular mechanisms by which these complex regulatory systems become malfunctional at specific stages of AD development and progression, which will facilitate future development of novel therapeutic interventions. It is also critical to investigate all key components of the protein degradation pathways, both upstream and downstream, to improve our abilities to manipulate transport pathways with higher efficacy and less side effects.


Assuntos
Doença de Alzheimer/metabolismo , Endossomos/fisiologia , Lisossomos/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Ubiquitina/fisiologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia , Humanos , Transdução de Sinais , Proteínas tau/metabolismo
19.
Neurobiol Learn Mem ; 165: 106791, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-29398581

RESUMO

The ubiquitin proteasome system (UPS) is a highly conserved pathway that tightly regulates protein turnover in cells. This process is integral to neuronal development, differentiation, and function. Several members of the UPS are disrupted in neuropsychiatric disorders, highlighting the importance of this pathway in brain development and function. In this review, we discuss some of these pathway members, the molecular processes they regulate, and the potential for targeting the UPS in an effort to develop therapeutic strategies in neuropsychiatric and neurodevelopmental disorders.


Assuntos
Transtornos Mentais/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Transtorno do Espectro Autista/metabolismo , Encefalopatias/metabolismo , Humanos , Deficiência Intelectual/metabolismo , Redes e Vias Metabólicas/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Esquizofrenia/metabolismo , Ubiquitina/fisiologia
20.
Adv Exp Med Biol ; 1088: 347-368, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30390260

RESUMO

Muscle atrophy in aging is characterized by progressive loss of muscle mass and function. Muscle mass is determined by the balance of synthesis and degradation of protein, which are regulated by several signaling pathways such as ubiquitin-proteasome system, autophagy-lysosome systems, oxidative stress, proinflammatory cytokines, hormones, and so on. Sufficient nutrition can enhance protein synthesis, while exercise can improve the quality of life in the elderly. This chapter will discuss the epidemiology, pathogenesis, as well as the current treatment for aging-induced muscular atrophy.


Assuntos
Envelhecimento/patologia , Músculo Esquelético/patologia , Atrofia Muscular/fisiopatologia , Autofagia , Citocinas/fisiologia , Humanos , Proteínas Musculares , Atrofia Muscular/epidemiologia , Atrofia Muscular/terapia , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/fisiologia , Transdução de Sinais , Ubiquitina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...