Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.223
Filtrar
1.
Int J Biol Sci ; 20(13): 5070-5086, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39430237

RESUMO

Peroxiredoxin 1 (PRDX1) is a potent antioxidant protein that displays a unique molecular chaperone activity. However, the role of overexpression of PRDX1 in colorectal cancer (CRC) was elusive. Herein, we found that the number of AOM/DSS-induced colitis-associated CRC in PRDX1 knockout mice was significantly lower than that in wild-type mice, concomitant with the downregulation of NRF2 and GPX4. Mechanistically, RNA sequencing results indicated that knockdown of PRDX1 resulted in a significant reduction of NRF2, which further triggered ROS-induced mitochondrial dysfunction and lipid peroxidation-induced ferroptosis in CRC cells. Notably, PRDX1 inhibited NRF2 degradation and promoted NRF2 nuclear translocation, thereby triggering the transcription of GPX4. Immunoprecipitation-mass spectrometry (IP-MS) and Co-immunoprecipitation (Co-IP) assays revealed that PRDX1 could act as a molecular chaperone by binding to CUL3 to inhibit NRF2 ubiquitination. Importantly, the binding of PRDX1 to CUL3 was enhanced by conoidin A but abolished by the PRDX1 Cys83Ser mutant. The inhibitory effects of PRDX1 knockdown on CRC could be attenuated by NRF2 activation or ferrostatin-1 administration in vivo. Collectively, these results provide a novel insight into the molecular chaperone activity of PRDX1 in promoting CRC progression through suppression of CUL3-mediated NRF2 degradation, suggesting PRDX1 Cys83 is a potential drug target in inhibiting CRC.


Assuntos
Neoplasias Colorretais , Ferroptose , Camundongos Knockout , Fator 2 Relacionado a NF-E2 , Peroxirredoxinas , Animais , Humanos , Masculino , Camundongos , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas Culina/metabolismo , Camundongos Endogâmicos C57BL , Chaperonas Moleculares/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Peroxirredoxinas/metabolismo , Peroxirredoxinas/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo
2.
Biomolecules ; 14(10)2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39456142

RESUMO

Ubiquitination is one of the most important post-translational modifications in eukaryotes. The ubiquitination cascade includes ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3). The E3 ligases, responsible for substrate recognition, are the most abundant and varied proteins in the cascade and the most studied. SKP1-CUL1-F-Box (SCF)-type E3 ubiquitin ligases are multi-subunit RING (Really Interesting New Gene) E3 ubiquitin ligases, composed of CUL1 (Cullin 1), RBX1 (RING BOX 1), SKP1 (S-phase Kinase-associated Protein 1), and F-box proteins. In vitro ubiquitination assays, used for studying the specific recognition of substrate proteins by E3 ubiquitin ligases, require the purification of all components involved in the cascade, and for assays with SCF-type E3 ligases, additional proteins (several SCF complex subunits). Here, the Duet expression system was used to co-express E1, E2, ubiquitin, ubiquitylation target (substrate), and the four subunits of a SCF-type E3 ligase in E. coli. When these proteins co-exist in bacterial cells, ubiquitination occurs and can be detected by Western Blot. The effectiveness of this bacterial system for detecting ubiquitination cascade activity was demonstrated by replicating both AtSCFTIR1-mediated and human SCFFBXO28-mediated ubiquitylation in bacteria. This system provides a basic but adaptable platform for the study of SCF-type E3 ubiquitin ligases.


Assuntos
Escherichia coli , Proteínas Ligases SKP Culina F-Box , Ubiquitinação , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Ubiquitina-Proteína Ligases/metabolismo , Humanos , Proteínas Culina/metabolismo
3.
Cells ; 13(19)2024 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-39404389

RESUMO

Mdmx (Mdm4) is established as an oncogene mainly through repression of the p53 tumour suppressor. On the other hand, anti-oncogenic functions for Mdmx have also been proposed, but the underlying regulatory pathways remain unknown. Investigations into the effect of inhibitors for the NEDD8 pathway in p53 activation, human cell morphology, and in cell motility during gastrulation in Xenopus embryos revealed an anti-invasive function of Mdmx. Through stabilisation and activation of the RhoA GTPase, Mdmx is required for the anti-invasive effects of NEDDylation inhibitors. Mechanistically, through its Zn finger domain, Mdmx preferentially interacts with the inactive GDP-form of RhoA. This protects RhoA from degradation and allows for RhoA targeting to the plasma membrane for its subsequent activation. The effect is transient, as prolonged NEDDylation inhibition targets Mdmx for degradation, which subsequently leads to RhoA destabilisation. Surprisingly, Mdmx degradation requires non-NEDDylated (inactive) Culin4A and the Mdm2 E3-ligase. This study reveals that Mdmx can control cell invasion through RhoA stabilisation/activation, which is potentially linked to the reported anti-oncogenic functions of Mdmx. As inhibitors of the NEDD8 pathway are in clinical trials, the status of Mdmx may be a critical determinant for the anti-tumour effects of these inhibitors.


Assuntos
Proteína NEDD8 , Proteína rhoA de Ligação ao GTP , Humanos , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína NEDD8/metabolismo , Animais , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Pirimidinas/farmacologia , Proteínas de Ciclo Celular/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Culina/metabolismo , Invasividade Neoplásica , Gastrulação , Proteólise/efeitos dos fármacos , Ciclopentanos/farmacologia , Ciclopentanos/metabolismo
4.
Cells ; 13(19)2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39404417

RESUMO

The ongoing obesity epidemic has raised awareness of the complex physiology of adipose tissue. Abnormal adipocyte differentiation results in the development of systemic metabolic disorders such as insulin resistance and diabetes. The conjugation of NEDD8 (neural precursor cell expressed, developmentally downregulated 8) to target protein, termed neddylation, has been shown to mediate adipogenesis. However, much remains unknown about its role in adipogenesis. Here, we demonstrated that neddylation and its targets, the cullin (CUL) family members, are differentially regulated during mouse and human adipogenesis. Inhibition of neddylation by MLN4924 significantly reduced adipogenesis of 3T3-L1 and human stromal vascular cells. Deletion of NAE1, a subunit of the only NEDD8 E1 enzyme, suppressed neddylation and impaired adipogenesis. Neddylation deficiency did not affect mitotic cell expansion. Instead, it disrupted CREB/CEBPß/PPARγ signaling, essential for adipogenesis. Interestingly, among the neddylation-targeted CUL family members, deletion of CUL3, but not CUL1, CUL2, or CUL4A, largely replicated the adipogenic defects observed with neddylation deficiency. A PPARγ agonist minimally rescued the adipogenic defects caused by the deletion of NAE1 and CUL3. In conclusion, our study demonstrates that neddylation and its targeted CUL3 are crucial for adipogenesis. These findings provide potential targets for therapeutic intervention in obesity and metabolic disorders.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Diferenciação Celular , Proteínas Culina , Proteína NEDD8 , Proteínas Culina/metabolismo , Adipócitos/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Humanos , Camundongos , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Proteína NEDD8/metabolismo , Proteína NEDD8/genética , Diferenciação Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , PPAR gama/metabolismo , Ciclopentanos/farmacologia , Pirimidinas/farmacologia , Enzimas Ativadoras de Ubiquitina
5.
Funct Integr Genomics ; 24(6): 204, 2024 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-39476129

RESUMO

This study aimed to explore the underlying mechanism of neddylation in macrophage polarization during sepsis. A mouse model of sepsis was established by cecal ligation and puncture (CLP). ELISA and Flow cytometry were performed to analyze the generation of pro-inflammatory factors and M1/M2 macrophage polarization, respectively. Western blotting was applied to detect NEDD8-mediated neddylation and glycolysis-related proteins. ECAR method was used to analyze the glycolysis level. HE staining was applied to detect the lung injury. The bacterial load in peritoneal cavity and peripheral blood was determined by counting the colony-forming units. The results showed the upregulated neddylation, M1 polarization and glycolysis of macrophage in patients with sepsis and CLP-challenged mice. NEDD8-mediated Cullin1 neddylation promoted M1 polarization and glycolysis to accelerate inflammation via NF-κB p65 pathway in E.coli-treated Raw264.7 cells. MLN4924 treatment alleviated sepsis by inhibiting neddylation to prevent M1 polarization in CLP-challenged mice. In summary, this study demonstrated that upregulation of NEDD8-mediated Cullin1 neddylation promotes glycolysis and M1 polarization of macrophage via NF-κB p65 pathway, accelerating inflammation in sepsis.


Assuntos
Proteínas Culina , Glicólise , Macrófagos , Proteína NEDD8 , Sepse , Fator de Transcrição RelA , Animais , Proteínas Culina/metabolismo , Proteínas Culina/genética , Sepse/metabolismo , Sepse/genética , Camundongos , Proteína NEDD8/metabolismo , Proteína NEDD8/genética , Macrófagos/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/genética , Células RAW 264.7 , Humanos , Regulação para Cima , Transdução de Sinais , Masculino , Camundongos Endogâmicos C57BL , Ciclopentanos/farmacologia , Pirimidinas/farmacologia , Inflamação/metabolismo , Inflamação/genética
6.
Biochem Biophys Res Commun ; 735: 150811, 2024 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-39406020

RESUMO

Cullin-RING E3 ubiquitin ligases (CRLs) constitute the largest family of ubiquitin ligase and play important roles in regulation of proteostasis. Here we presented the cryo-EM structure of CRL1FBXO4, a member of Cullin-1 E3 ligase. CRL1FBXO4 adopts a homodimer architecture. Structural analysis revealed that in the CRL1FBXO4 protomer, the substrate recognition subunit FBXO4 interacts both the adaptor protein SKP1, and the scaffold protein CUL1 via hydrophobic and electrostatic interactions. Two FBXO4 forms a domain-swapped dimer in the CRL1FBXO4 structure, which constitutes the basis for the dimerization of CRL1FBXO4. Inspired by the cryo-EM density, we modeled the architecture of whole CRL1FBXO4 as a symmetrical dimer, which provides insights into CRL1FBXO4-medaited turnover of oncogene proteins.


Assuntos
Proteínas Culina , Proteínas F-Box , Modelos Moleculares , Multimerização Proteica , Proteínas Culina/metabolismo , Proteínas Culina/química , Humanos , Proteínas F-Box/metabolismo , Proteínas F-Box/química , Microscopia Crioeletrônica , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/química , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteínas Quinases Associadas a Fase S/química , Proteínas Quinases Associadas a Fase S/genética , Ligação Proteica , Conformação Proteica , Proteínas de Transporte
7.
Mol Biol (Mosk) ; 58(2): 305-313, 2024.
Artigo em Russo | MEDLINE | ID: mdl-39355887

RESUMO

An RNA interference-based method was proposed to achieve an inducible knockdown of genes essential for cell viability. In the method, a genetic cassette in which a copper ion-dependent inducible metallothionein promoter controls expression of a siRNA precursor is inserted into a genomic pre-integrated transgene by CRIPSR/Cas9 technology. The endogenous siRNA source allows the gene knockdown in cell cultures that are refractory to conventional transfection with exogenous siRNA. The efficiency of the method was demonstrated in Drosophila ovarian somatic cell culture (OSC) for two genes that are essential for oogenesis: Cul3, encoding a component of the multiprotein ubiquitin-ligase complex with versatile functions in proteostasis, and cut, encoding a transcription factor regulating differentiation of ovarian follicular cells.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Animais , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Proteínas Culina/genética , Proteínas Culina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ovário/metabolismo , Ovário/citologia , Oogênese/genética , Interferência de RNA , Genes Essenciais , Sistemas CRISPR-Cas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
8.
Stem Cell Reports ; 19(10): 1489-1504, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39270650

RESUMO

Translating genetic findings for neurodevelopmental and psychiatric disorders (NPDs) into actionable disease biology would benefit from large-scale and unbiased functional studies of NPD genes. Leveraging the cytosine base editing (CBE) system, we developed a pipeline for clonal loss-of-function (LoF) allele mutagenesis in human induced pluripotent stem cells (hiPSCs) by introducing premature stop codons (iSTOP) that lead to mRNA nonsense-mediated decay (NMD) or protein truncation. We tested the pipeline for 23 NPD genes on 3 hiPSC lines and achieved highly reproducible, efficient iSTOP editing in 22 genes. Using RNA sequencing (RNA-seq), we confirmed their pluripotency, absence of chromosomal abnormalities, and NMD. Despite high editing efficiency, three schizophrenia risk genes (SETD1A, TRIO, and CUL1) only had heterozygous LoF alleles, suggesting their essential roles for cell growth. We found that CUL1-LoF reduced neurite branches and synaptic puncta density. This iSTOP pipeline enables a scaled and efficient LoF mutagenesis of NPD genes, yielding an invaluable shareable resource.


Assuntos
Alelos , Células-Tronco Pluripotentes Induzidas , Mutação com Perda de Função , Transtornos do Neurodesenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Transtornos do Neurodesenvolvimento/genética , Transtornos Mentais/genética , Edição de Genes , Degradação do RNAm Mediada por Códon sem Sentido , Mutagênese , Códon sem Sentido , Predisposição Genética para Doença , Sistemas CRISPR-Cas , Proteínas Culina/genética
9.
Mol Med Rep ; 30(5)2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39239747

RESUMO

Cholangiocarcinoma (CCA) is an extremely aggressive malignancy arising from the epithelial cells lining the bile ducts. It presents a substantial global health issue, with the highest incidence rates, ranging from 40­100 cases/100,000 individuals, found in Southeast Asia, where liver fluke infection is endemic. In Europe and America, incidence rates range from 0.4­2 cases/100,000 individuals. Globally, mortality rates range from 0.2­2 deaths/100,000 person­years and are increasing in most countries. Chemotherapy is the primary treatment for advanced CCA due to limited options from late­stage diagnosis, but its efficacy is hindered by drug­resistant phenotypes. In a previous study, proteomics analysis of drug­resistant CCA cell lines (KKU­213A­FR and KKU­213A­GR) and the parental KKU­213A line identified cullin 3 (Cul3) as markedly overexpressed in drug­resistant cells. Cul3, a scaffold protein within CUL3­RING ubiquitin ligase complexes, is crucial for ubiquitination and proteasome degradation, yet its role in drug­resistant CCA remains to be elucidated. The present study aimed to elucidate the role of Cul3 in drug­resistant CCA cell lines. Reverse transcription­quantitative PCR and western blot analyses confirmed significantly elevated Cul3 mRNA and protein levels in drug­resistant cell lines compared with the parental control. Short interfering RNA­mediated Cul3 knockdown sensitized cells to 5­fluorouracil and gemcitabine and inhibited cell proliferation, colony formation, migration and invasion. In addition, Cul3 knockdown induced G0/G1 cell cycle arrest and suppressed key cell cycle regulatory proteins, cyclin D, cyclin­dependent kinase (CDK)4 and CDK6. Bioinformatics analysis of CCA patient samples using The Cancer Genome Atlas data revealed Cul3 upregulation in CCA tissues compared with normal bile duct tissues. STRING analysis of upregulated proteins in drug­resistant CCA cell lines identified a highly interactive Cul3 network, including COMM Domain Containing 3, Ariadne RBR E3 ubiquitin protein ligase 1, Egl nine homolog 1, Proteasome 26S Subunit Non­ATPase 13, DExH­box helicase 9 and small nuclear ribonucleoprotein polypeptide G, which showed a positive correlation with Cul3 in CCA tissues. Knocking down Cul3 significantly suppressed the mRNA expression of these genes, suggesting that Cul3 may act as an upstream regulator of them. Gene Ontology analysis revealed that the majority of these genes were categorized under binding function, metabolic process, cellular anatomical entity, protein­containing complex and protein­modifying enzyme. Taken together, these findings highlighted the biological and clinical significance of Cul3 in drug resistance and progression of CCA.


Assuntos
Neoplasias dos Ductos Biliares , Proliferação de Células , Colangiocarcinoma , Proteínas Culina , Resistencia a Medicamentos Antineoplásicos , Humanos , Colangiocarcinoma/metabolismo , Colangiocarcinoma/genética , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/patologia , Proteínas Culina/metabolismo , Proteínas Culina/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Fenótipo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Gencitabina , Movimento Celular/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Antineoplásicos/farmacologia
10.
Theranostics ; 14(13): 4916-4932, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39267786

RESUMO

Rationale: Photoreceptor loss is a primary pathological feature of retinal degeneration (RD) with limited treatment strategies. RNA interference (RNAi) has emerged as a promising method of gene therapy in regenerative medicine. However, the transfer of RNAi therapeutics to photoreceptors and the deficiency of effective therapeutic targets are still major challenges in the treatment of RD. Methods: In this study, photoreceptor-derived extracellular vesicles (PEVs) conjugated with photoreceptor-binding peptide MH42 (PEVsMH42) were prepared using the anchoring peptide CP05. Transcriptome sequencing was applied to investigate the potential therapeutic target of RD. We then engineered PEVsMH42 with specific small-interfering RNAs (siRNAs) through electroporation and evaluated their therapeutic efficacy in N-methyl-N-nitrosourea (MNU)-induced RD mice and Pde6ßrd1/rd1 mutant mice. Results: PEVsMH42 were selectively accumulated in photoreceptors after intravitreal injection. Cullin-7 (Cul7) was identified as a novel therapeutic target of RD. Taking advantage of the established PEVsMH42, siRNAs targeting Cul7 (siCul7) were efficiently delivered to photoreceptors and consequently blocked the expression of Cul7. Moreover, suppression of Cul7 effectively protected photoreceptors to alleviate RD both in MNU-induced mouse model and Pde6ßrd1/rd1 mutant mouse model. Mechanistically, PEVsMH42 loaded with siCul7 (PEVsMH42-siCul7)-induced Cul7 downregulation was responsible for preventing Cul7-mediated glutathione peroxidase 4 (Gpx4) ubiquitination and degradation, resulting in the inhibition of photoreceptor ferroptosis. Conclusions: In summary, PEVsMH42-siCul7 attenuate photoreceptor ferroptosis to treat RD by inhibiting Cul7-induced ubiquitination of Gpx4. Our study develops a PEVs-based platform for photoreceptor-targeted delivery and highlights the potential of PEVsMH42-siCul7 as effective therapeutics for RD.


Assuntos
Vesículas Extracelulares , RNA Interferente Pequeno , Degeneração Retiniana , Animais , Degeneração Retiniana/terapia , Degeneração Retiniana/genética , Camundongos , Vesículas Extracelulares/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/administração & dosagem , Proteínas Culina/genética , Proteínas Culina/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Terapia Genética/métodos , Células Fotorreceptoras/metabolismo , Humanos , Células Fotorreceptoras de Vertebrados/metabolismo , Injeções Intravítreas , Metilnitrosoureia
11.
Cells ; 13(17)2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39273066

RESUMO

Many cellular processes are regulated by proteasome-mediated protein degradation, including regulation of signaling pathways and gene expression. Among the pathways regulated by the ubiquitin-proteasome system is the Hedgehog pathway and its downstream effectors, the Gli transcription factors. Here we provide evidence that proteasomal activity is necessary for maintaining the activation of the Hedgehog pathway, and this crucial event takes place at the level of Gli proteins. We undertook extensive work to demonstrate the specificity of the observed phenomenon by ruling out the involvement of primary cilium, impaired nuclear import, failed dissociation from Sufu, microtubule stabilization, and stabilization of Gli repressor forms. Moreover, we showed that proteasomal-inhibition-mediated Hedgehog pathway downregulation is not restricted to the NIH-3T3 cell line. We demonstrated, using CRISPR/Ca9 mutagenesis, that neither Gli1, Gli2, nor Gli3 are solely responsible for the Hedgehog pathway downregulation upon proteasome inhibitor treatment, and that Cul3 KO renders the same phenotype. Finally, we report two novel E3 ubiquitin ligases, Btbd9 and Kctd3, known Cul3 interactors, as positive Hedgehog pathway regulators. Our data pave the way for a better understanding of the regulation of gene expression and the Hedgehog signaling pathway.


Assuntos
Proteínas Culina , Proteínas Hedgehog , Complexo de Endopeptidases do Proteassoma , Transdução de Sinais , Ubiquitinação , Animais , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Camundongos , Células NIH 3T3 , Proteínas Culina/metabolismo , Proteínas Culina/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Humanos , Regulação da Expressão Gênica
12.
mBio ; 15(10): e0167224, 2024 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-39207167

RESUMO

Malaria is a mosquito-borne infectious disease caused by unicellular eukaryotic parasites of the Plasmodium genus. Protein ubiquitination by E3 ligases is a critical post-translational modification required for various cellular processes during the lifecycle of Plasmodium parasites. However, little is known about the repertoire and function of these enzymes in Plasmodium. Here, we show that Plasmodium expresses a conserved cullin RING E3 ligase (CRL) complex that is functionally related to CRL4 in other eukaryotes. In P. falciparum asexual blood stages, a cullin-4 scaffold interacts with the RING protein RBX1, the adaptor protein DDB1, and a set of putative receptor proteins that may determine substrate specificity for ubiquitination. These receptor proteins contain WD40-repeat domains and include WD-repeat protein important for gametogenesis 1 (WIG1). This CRL4-related complex is also expressed in P. berghei gametocytes, with WIG1 being the only putative receptor detected in both the schizont and gametocyte stages. WIG1 disruption leads to a complete block in microgamete formation. Proteomic analyses indicate that WIG1 disruption alters proteostasis of ciliary proteins and components of the DNA replication machinery during gametocytogenesis. Further analysis by ultrastructure expansion microscopy (U-ExM) indicates that WIG1-dependent depletion of ciliary proteins is associated with impaired the formation of the microtubule organization centers that coordinate mitosis with axoneme formation and altered DNA replication during microgametogenesis. This work identifies a CRL4-related ubiquitin ligase in Plasmodium that is critical for the formation of microgametes by regulating proteostasis of ciliary and DNA replication proteins.IMPORTANCEPlasmodium parasites undergo fascinating lifecycles with multiple developmental steps, converting into morphologically distinct forms in both their mammalian and mosquito hosts. Protein ubiquitination by ubiquitin ligases emerges as an important post-translational modification required to control multiple developmental stages in Plasmodium. Here, we identify a cullin RING E3 ubiquitin ligase (CRL) complex expressed in the replicating asexual blood stages and in the gametocyte stages that mediate transmission to the mosquito. WIG1, a putative substrate recognition protein of this ligase complex, is essential for the maturation of microgametocytes into microgametes upon ingestion by a mosquito. More specifically, WIG1 is required for proteostasis of ciliary proteins and components of the DNA replication machinery during gametocytogenesis. This requirement is linked to DNA replication and microtubule organization center formation, both critical to the development of flagellated microgametes.


Assuntos
Plasmodium falciparum , Proteínas de Protozoários , Ubiquitina-Proteína Ligases , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Plasmodium falciparum/genética , Plasmodium falciparum/enzimologia , Plasmodium falciparum/metabolismo , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium berghei/genética , Plasmodium berghei/enzimologia , Plasmodium berghei/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Microtúbulos/metabolismo , Animais , Ubiquitinação , Humanos , Proteínas Culina/metabolismo , Proteínas Culina/genética
13.
Kidney360 ; 5(9): 1386-1393, 2024 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-39120943

RESUMO

The sodium chloride cotransporter (NCC) fine-tunes Na + balance and indirectly affects the homeostasis of other ions including K + , Mg 2+ , and Ca 2+ . Owing to its effects on Na + balance, BP is significantly affected by alterations in NCC activity. Several factors have been reported to influence the expression and activity of NCC. One critical factor is NCC phosphorylation/dephosphorylation that occurs at key serine-threonine amino acid residues of the protein. Phosphorylation, which results in increased NCC activity, is mediated by the with no lysine [K] (WNK)-SPS-related proline alanine rich kinase (SPAK)/OSR1 kinases. NCC activation stimulates reabsorption of Na + , increasing extracellular fluid volume and hence BP. On the other hand, proteasomal degradation of WNK kinases after ubiquitination by the Cullin 3-Kelch-like 3 E3 ubiquitin ligase complex and dephosphorylation pathways oppose WNK-SPAK/OSR1-mediated NCC activation. Components of the Cullin 3/Kelch-like 3-WNK-SPAK/OSR1 regulatory pathway may be targets for novel antihypertensive drugs. In this review, we outline the impact of these regulators on the activity of NCC and the consequent effect on BP.


Assuntos
Proteínas Culina , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Proteínas Serina-Treonina Quinases/metabolismo , Humanos , Animais , Proteínas Culina/metabolismo , Fosforilação , Pressão Sanguínea/fisiologia , Simportadores de Cloreto de Sódio/metabolismo , Simportadores de Cloreto de Sódio/genética , Proteína Quinase 1 Deficiente de Lisina WNK/metabolismo
14.
Am J Physiol Renal Physiol ; 327(4): F667-F682, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39205661

RESUMO

The disease familial hyperkalemic hypertension (FHHt; also known as Gordon syndrome) is caused by aberrant accumulation of with-no-lysine kinase (WNK4) activating the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) of the kidney. Mutations in cullin 3 (CUL3) cause FHHt by disrupting interaction with the deneddylase COP9 signalosome (CSN). Deletion of Cul3 or Jab1 (the catalytically active CSN subunit) along the entire nephron causes a partial FHHt phenotype with activation of the WNK4-STE20/SPS1-related proline/alanine-rich kinase (SPAK)-NCC pathway. However, progressive kidney injury likely prevents hypertension, hyperkalemia, and hyperchloremic metabolic acidosis associated with FHHt. We hypothesized that DCT-specific deletion would more closely model the disease. We used Slc12a3-Cre-ERT2 mice to delete Cul3 (DCT-Cul3-/-) or Jab1 (DCT-Jab1-/-) only in the DCT and examined the mice after short- and long-term deletion. Short-term DCT-specific knockout of both Cul3 and Jab1 mice caused elevated WNK4, pSPAKS373, and pNCCT53 abundance. However, neither model demonstrated changes in plasma K+, Cl-, or total CO2, even though no injury was present. Long-term DCT-Jab1-/- mice showed significantly lower NCC and parvalbumin abundance and a higher abundance of kidney injury molecule-1, a marker of proximal tubule injury. No injury or reduction in NCC or parvalbumin was observed in long-term DCT-Cul3-/- mice. In summary, the prevention of injury outside the DCT did not lead to a complete FHHt phenotype despite activation of the WNK4-SPAK-NCC pathway, possibly due to insufficient NCC activation. Chronically, only DCT-Jab1-/- mice developed tubule injury and atrophy of the DCT, suggesting a direct JAB1 effect or dysregulation of other cullins as mechanisms for injury.NEW & NOTEWORTHY CUL3 degrades WNK4, which prevents activation of NCC in the DCT. CSN regulation of CUL3 is impaired in the disease FHHt, causing accumulation of WNK4. Short-term DCT-specific disruption of CUL3 or the CSN in mice resulted in activation of the WNK4-SPAK-NCC pathway but not hyperkalemic metabolic acidosis found in FHHt. Tubule injury was observed only after long-term CSN disruption. The data suggest that disruption of other cullins may be the cause for the injury.


Assuntos
Complexo do Signalossomo COP9 , Proteínas Culina , Túbulos Renais Distais , Camundongos Knockout , Animais , Complexo do Signalossomo COP9/metabolismo , Complexo do Signalossomo COP9/genética , Proteínas Culina/metabolismo , Proteínas Culina/genética , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/patologia , Modelos Animais de Doenças , Camundongos , Pseudo-Hipoaldosteronismo/genética , Pseudo-Hipoaldosteronismo/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/genética , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/genética
15.
Clin Mol Hepatol ; 30(4): 895-913, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39098817

RESUMO

BACKGROUND/AIMS: Potassium channel tetramerization domain containing 17 (KCTD17) protein, an adaptor for the cullin3 (Cul3) ubiquitin ligase complex, has been implicated in various human diseases; however, its role in hepatocellular carcinoma (HCC) remains elusive. Here, we aimed to elucidate the clinical features of KCTD17, and investigate the mechanisms by which KCTD17 affects HCC progression. METHODS: We analyzed transcriptomic data from patients with HCC. Hepatocyte-specific KCTD17 deficient mice were treated with diethylnitrosamine (DEN) to assess its effect on HCC progression. Additionally, we tested KCTD17-directed antisense oligonucleotides for their therapeutic potential in vivo. RESULTS: Our investigation revealed the upregulation of KCTD17 expression in both tumors from patients with HCC and mouse models of HCC, in comparison to non-tumor controls. We identified the leucine zipper-like transcriptional regulator 1 (Lztr1) protein, a previously identified Ras destabilizer, as a substrate for KCTD17-Cul3 complex. KCTD17-mediated Lztr1 degradation led to Ras stabilization, resulting in increased proliferation, migration, and wound healing in liver cancer cells. Hepatocyte-specific KCTD17 deficient mice or liver cancer xenograft models were less susceptible to carcinogenesis or tumor growth. Similarly, treatment with KCTD17-directed antisense oligonucleotides (ASO) in a mouse model of HCC markedly lowered tumor volume as well as Ras protein levels, compared to those in control ASO-treated mice. CONCLUSION: KCTD17 induces the stabilization of Ras and downstream signaling pathways and HCC progression and may represent a novel therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular , Progressão da Doença , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Camundongos , Proteínas Culina/metabolismo , Proteínas Culina/genética , Proteínas ras/metabolismo , Proteínas ras/genética , Proliferação de Células , Masculino , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Dietilnitrosamina/toxicidade , Oligonucleotídeos Antissenso/metabolismo
16.
ACS Chem Biol ; 19(9): 1942-1952, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39194017

RESUMO

The RAS family of GTPases is among the most frequently mutated proteins in human cancer, creating a high clinical demand for therapies that counteract their signaling activity. An important layer of regulation that could be therapeutically exploited is the proteostatic regulation of the main RAS GTPases KRAS, NRAS, and HRAS, as well as the closely related members, MRAS and RIT1, by the leucine zipper-like transcriptional regulator 1 cullin 3 RING E3 ubiquitin ligase complex (CUL3LZTR1). Genetic inactivation of LZTR1, as observed in different cancer entities and Noonan syndrome leads to enhanced RAS GTPase abundance and altered MAPK pathway activation state. Novel therapeutic approaches to interfere with hyperactive RAS signaling, thereby complementing existing treatments, are highly sought after. Motivated by the growing arsenal of molecular glue degraders, we report the identification of novel chemical fragments that enhance the protein-protein interaction (PPI) of the KRAS-LZTR1 complex. We established a split-luciferase-based reporter assay that monitors the RAS GTPase-LZTR1 interaction in a scalable format, capable of capturing chemical, as well as mutational perturbations. Using this screening system, in combination with a small fragment library, we identified two fragments, C53 and Z86, that enhance the interaction of the KRAS-LZTR1 complex in a dose-dependent manner. Further orthogonal validation experiments using proximity biotinylation (BioID), thermal shift assays, and NMR spectroscopy demonstrated fragment-dependent enhanced recruitment of endogenous LZTR1 and physical engagement of KRAS. The two fragments, which potentiate the KRAS-LZTR1 interaction, serve as starting points for fragment-based drug discovery. Additionally, the assay we introduced is amenable to high-throughput screening to further explore the pharmacological modulation of the CUL3LZTR1-RAS GTPase complex.


Assuntos
Proteínas Proto-Oncogênicas p21(ras) , Humanos , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química , Proteínas Culina/metabolismo , Proteínas Culina/genética , Transativadores/metabolismo , Transativadores/genética , Transativadores/química , Ligação Proteica , Células HEK293 , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética
17.
Biomolecules ; 14(8)2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39199272

RESUMO

Mouse double minute 2 (MDM2) is an oncoprotein that is frequently overexpressed in tumors and enhances cellular transformation. Owing to the important role of MDM2 in modulating p53 function, it is crucial to understand the mechanism underlying the regulation of MDM2 levels. We identified ribosomal protein S4X-linked (RPS4X) as a novel binding partner of MDM2 and showed that RPS4X promotes MDM2 stability. RPS4X suppressed polyubiquitination of MDM2 by suppressing homodimer formation and preventing auto-ubiquitination. Moreover, RPS4X inhibited the interaction between MDM2 and Cullin1, a scaffold protein of the Skp1-Cullin1-F-box protein (SCF) complex and an E3 ubiquitin ligase for MDM2. RPS4X expression in cells enhanced the steady-state level of MDM2 protein. RPS4X was associated not only with MDM2 but also with Cullin1 and then blocked the MDM2/Cullin1 interaction. This is the first report of an interaction between ribosomal proteins (RPs) and Cullin1. Our results contribute to the elucidation of the MDM2 stabilization mechanism in cancer cells, expanding our understanding of the new functions of RPs.


Assuntos
Proteínas Culina , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Ribossômicas , Ubiquitinação , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Ribossômicas/metabolismo , Proteínas Ribossômicas/genética , Humanos , Proteínas Culina/metabolismo , Proteínas Culina/genética , Animais , Estabilidade Proteica , Camundongos , Ligação Proteica , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Células HEK293
19.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39145933

RESUMO

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Assuntos
Proteínas Culina , Interferon Tipo I , Proteólise , Fator de Transcrição STAT2 , Transdução de Sinais , Ubiquitina-Proteína Ligases , Ubiquitinação , Proteínas não Estruturais Virais , Infecção por Zika virus , Zika virus , Humanos , Fator de Transcrição STAT2/metabolismo , Zika virus/imunologia , Zika virus/fisiologia , Zika virus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Interferon Tipo I/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Proteínas Culina/metabolismo , Células A549 , Células HEK293 , Sistemas CRISPR-Cas
20.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189169, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-39117093

RESUMO

Cullin-RING ligase 4 (CRL4) has attracted enormous attentions because of its extensive regulatory roles in a wide variety of biological and pathological events, especially cancer-associated events. CRL4 exerts pleiotropic effects by targeting various substrates for proteasomal degradation or changes in activity through different internal compositions to regulate diverse events in cancer progression. In this review, we summarize the structure of CRL4 with manifold compositional modes and clarify the emerging functions and molecular mechanisms of CRL4 in a series of cancer-associated events.


Assuntos
Neoplasias , Humanos , Neoplasias/patologia , Neoplasias/enzimologia , Neoplasias/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Ubiquitinação , Proteínas Culina/metabolismo , Receptores de Interleucina-17
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...