Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 83(6): 927-941.e8, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36898370

RESUMO

Mitophagy is a form of selective autophagy that disposes of superfluous and potentially damage-inducing organelles in a tightly controlled manner. While the machinery involved in mitophagy induction is well known, the regulation of the components is less clear. Here, we demonstrate that TNIP1 knockout in HeLa cells accelerates mitophagy rates and that ectopic TNIP1 negatively regulates the rate of mitophagy. These functions of TNIP1 depend on an evolutionarily conserved LIR motif as well as an AHD3 domain, which are required for binding to the LC3/GABARAP family of proteins and the autophagy receptor TAX1BP1, respectively. We further show that phosphorylation appears to regulate its association with the ULK1 complex member FIP200, allowing TNIP1 to compete with autophagy receptors, which provides a molecular rationale for its inhibitory function during mitophagy. Taken together, our findings describe TNIP1 as a negative regulator of mitophagy that acts at the early steps of autophagosome biogenesis.


Assuntos
Proteínas Relacionadas à Autofagia , Autofagia , Mitofagia , Humanos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/genética , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitofagia/genética , Proteínas de Neoplasias/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(25): e2322588121, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38861598

RESUMO

The nematode intestine is the primary site for nutrient uptake and storage as well as the synthesis of biomolecules; lysosome-related organelles known as gut granules are important for many of these functions. Aspects of intestine biology are not well understood, including the export of the nutrients it imports and the molecules it synthesizes, as well as the complete functions and protein content of the gut granules. Here, we report a mass spectrometry (MS)-based proteomic analysis of the intestine of the Caenorhabditis elegans and of its gut granules. Overall, we identified approximately 5,000 proteins each in the intestine and the gonad and showed that most of these proteins can be detected in samples extracted from a single worm, suggesting the feasibility of individual-level genetic analysis using proteomes. Comparing proteomes and published transcriptomes of the intestine and the gonad, we identified proteins that appear to be synthesized in the intestine and then transferred to the gonad. To identify gut granule proteins, we compared the proteome of individual intestines deficient in gut granules to the wild type. The identified gut granule proteome includes proteins known to be exclusively localized to the granules and additional putative gut granule proteins. We selected two of these putative gut granule proteins for validation via immunohistochemistry, and our successful confirmation of both suggests that our strategy was effective in identifying the gut granule proteome. Our results demonstrate the practicability of single-tissue MS-based proteomic analysis in small organisms and in its future utility.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Lisossomos , Proteômica , Animais , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteômica/métodos , Lisossomos/metabolismo , Proteoma/metabolismo , Intestinos , Mucosa Intestinal/metabolismo , Gônadas/metabolismo , Espectrometria de Massas/métodos , Organelas/metabolismo
3.
Am J Hum Genet ; 110(11): 1959-1975, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37883978

RESUMO

Valosin-containing protein (VCP) is an AAA+ ATPase that plays critical roles in multiple ubiquitin-dependent cellular processes. Dominant pathogenic variants in VCP are associated with adult-onset multisystem proteinopathy (MSP), which manifests as myopathy, bone disease, dementia, and/or motor neuron disease. Through GeneMatcher, we identified 13 unrelated individuals who harbor heterozygous VCP variants (12 de novo and 1 inherited) associated with a childhood-onset disorder characterized by developmental delay, intellectual disability, hypotonia, and macrocephaly. Trio exome sequencing or a multigene panel identified nine missense variants, two in-frame deletions, one frameshift, and one splicing variant. We performed in vitro functional studies and in silico modeling to investigate the impact of these variants on protein function. In contrast to MSP variants, most missense variants had decreased ATPase activity, and one caused hyperactivation. Other variants were predicted to cause haploinsufficiency, suggesting a loss-of-function mechanism. This cohort expands the spectrum of VCP-related disease to include neurodevelopmental disease presenting in childhood.


Assuntos
Doenças Musculares , Transtornos do Neurodesenvolvimento , Adulto , Humanos , Proteína com Valosina/genética , Hipotonia Muscular , Mutação de Sentido Incorreto/genética
4.
J Proteome Res ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38832920

RESUMO

The advancement of sophisticated instrumentation in mass spectrometry has catalyzed an in-depth exploration of complex proteomes. This exploration necessitates a nuanced balance in experimental design, particularly between quantitative precision and the enumeration of analytes detected. In bottom-up proteomics, a key challenge is that oversampling of abundant proteins can adversely affect the identification of a diverse array of unique proteins. This issue is especially pronounced in samples with limited analytes, such as small tissue biopsies or single-cell samples. Methods such as depletion and fractionation are suboptimal to reduce oversampling in single cell samples, and other improvements on LC and mass spectrometry technologies and methods have been developed to address the trade-off between precision and enumeration. We demonstrate that by using a monosubstrate protease for proteomic analysis of single-cell equivalent digest samples, an improvement in quantitative accuracy can be achieved, while maintaining high proteome coverage established by trypsin. This improvement is particularly vital for the field of single-cell proteomics, where single-cell samples with limited number of protein copies, especially in the context of low-abundance proteins, can benefit from considering analyte complexity. Considerations about analyte complexity, alongside chromatographic complexity, integration with data acquisition methods, and other factors such as those involving enzyme kinetics, will be crucial in the design of future single-cell workflows.

5.
BMC Bioinformatics ; 24(1): 239, 2023 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280522

RESUMO

BACKGROUND: The analysis of mass spectrometry-based quantitative proteomics data can be challenging given the variety of established analysis platforms, the differences in reporting formats, and a general lack of approachable standardized post-processing analyses such as sample group statistics, quantitative variation and even data filtering. We developed tidyproteomics to facilitate basic analysis, improve data interoperability and potentially ease the integration of new processing algorithms, mainly through the use of a simplified data-object. RESULTS: The R package tidyproteomics was developed as both a framework for standardizing quantitative proteomics data and a platform for analysis workflows, containing discrete functions that can be connected end-to-end, thus making it easier to define complex analyses by breaking them into small stepwise units. Additionally, as with any analysis workflow, choices made during analysis can have large impacts on the results and as such, tidyproteomics allows researchers to string each function together in any order, select from a variety of options and in some cases develop and incorporate custom algorithms. CONCLUSIONS: Tidyproteomics aims to simplify data exploration from multiple platforms, provide control over individual functions and analysis order, and serve as a tool to assemble complex repeatable processing workflows in a logical flow. Datasets in tidyproteomics are easy to work with, have a structure that allows for biological annotations to be added, and come with a framework for developing additional analysis tools. The consistent data structure and accessible analysis and plotting tools also offers a way for researchers to save time on mundane data manipulation tasks.


Assuntos
Proteômica , Software , Proteômica/métodos , Algoritmos , Espectrometria de Massas/métodos , Fluxo de Trabalho
6.
Mol Microbiol ; 118(4): 321-335, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36271736

RESUMO

Pseudomonas aeruginosa is an opportunistic bacterial pathogen that often encounters hypoxic/anoxic environments within the host, which increases its tolerance to many conventional antibiotics. Toward identifying novel treatments, we explored the therapeutic potential of chlorate, a pro-drug that kills hypoxic/anoxic, antibiotic-tolerant P. aeruginosa populations. While chlorate itself is relatively nontoxic, it is enzymatically reduced to the toxic oxidizing agent, chlorite, by hypoxically induced nitrate reductase. To better assess chlorate's therapeutic potential, we investigated mechanisms of chlorate toxicity and resistance in P. aeruginosa. We used transposon mutagenesis to identify genes that alter P. aeruginosa fitness during chlorate treatment, finding that methionine sulfoxide reductases (Msr), which repair oxidized methionine residues, support survival during chlorate stress. Chlorate treatment leads to proteome-wide methionine oxidation, which is exacerbated in a ∆msrA∆msrB strain. In response to chlorate, P. aeruginosa upregulates proteins involved in a wide range of functions, including metabolism, DNA replication/repair, protein repair, transcription, and translation, and these newly synthesized proteins are particularly vulnerable to methionine oxidation. The addition of exogenous methionine partially rescues P. aeruginosa survival during chlorate treatment, suggesting that widespread methionine oxidation contributes to death. Finally, we found that mutations that decrease nitrate reductase activity are a common mechanism of chlorate resistance.


Assuntos
Cloratos , Pró-Fármacos , Cloratos/metabolismo , Cloratos/farmacologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Metionina Sulfóxido Redutases/genética , Proteoma , Nitratos/metabolismo , Nitrato Redutase , Antibacterianos/farmacologia , Oxidantes , Metionina
7.
Bioorg Med Chem ; 62: 116726, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35364523

RESUMO

RUVBL1 and RUVBL2 are highly conserved AAA ATPases (ATPases Associated with various cellular Activities) and highly relevant to the progression of cancer, which makes them attractive targets for novel therapeutic anticancer drugs. In this work, docking-based virtual screening was performed to identify compounds with activity against the RUVBL1/2 complex. Seven compounds showed inhibitory activity against the complex in both enzymatic and cellular assays. A series of pyrazolo[1,5-a]pyrimidine-3-carboxamide analogs were synthesized based on the scaffold of compound 15 with inhibitory activity and good potential for structural manipulation. Analysis of the structure-activity relationship identified the benzyl group on R2 and aromatic ring-substituted piperazinyl on R4 as essential for inhibitory activity against the RUVBL1/2 complex. Of these, compound 18, which has IC50 values of 6.0 ± 0.6 µM and 7.7 ± 0.9 µM against RUVBL1/2 complex and RUVBL1 respectively, showed the most potent inhibition in cell lines A549, H1795, HCT116, and MDA-MB-231 with IC50 values of 15 ± 1.2 µM, 15 ± 1.8 µM, 11 ± 1.0 µM, and 8.9 ± 0.9 µM respectively. A docking study of the compound was performed to predict the binding mode of pyrazolo[1,5-a]pyrimidine-3-carboxamides. Furthermore, mass spectrometry-based proteomic analysis was employed to explore cellular proteins dysregulated by treatment with compounds 16, 18, and 19. Together, the data from these analyses suggest that that compound 18 could serve as a starting point for structural modifications in order to improve potency, selectivity, and pharmacokinetic parameters of potential therapeutic molecules.


Assuntos
Adenosina Trifosfatases , Antineoplásicos , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Adenosina Trifosfatases/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Proteínas de Transporte/metabolismo , DNA Helicases , Ensaios de Seleção de Medicamentos Antitumorais , Proteômica , Relação Estrutura-Atividade
8.
Biochem J ; 478(12): 2309-2319, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34032266

RESUMO

Enzyme replacement therapy (ERT) is a scientifically rational and clinically proven treatment for lysosomal storage diseases. Most enzymes used for ERT are purified from the culture supernatant of mammalian cells. However, it is challenging to purify lysosomal enzymes with sufficient quality and quantity for clinical use due to their low secretion levels in mammalian cell systems. To improve the secretion efficiency of recombinant lysosomal enzymes, we evaluated the impact of artificial signal peptides on the production of recombinant lysosomal enzymes in Chinese hamster ovary (CHO) cell lines. We engineered two recombinant human lysosomal enzymes, N-acetyl-α-glucosaminidase (rhNAGLU) and glucosamine (N-acetyl)-6-sulfatase (rhGNS), by replacing their native signal peptides with nine different signal peptides derived from highly secretory proteins and expressed them in CHO K1 cells. When comparing the native signal peptides, we found that rhGNS was secreted into media at higher levels than rhNAGLU. The secretion of rhNAGLU and rhGNS can, however, be carefully controlled by altering signal peptides. The secretion of rhNAGLU was relatively higher with murine Igκ light chain and human chymotrypsinogen B1 signal peptides, whereas Igκ light chain signal peptide 1 and human chymotrypsinogen B1 signal peptides were more effective for rhGNS secretion, suggesting that human chymotrypsinogen B1 signal peptide is the most appropriate for increasing lysosomal enzyme secretion. Collectively, our results indicate that altering signal peptide can modulate the secretion of recombinant lysosome enzymes and will enable lysosomal enzyme production for clinical use.


Assuntos
Acetilglucosaminidase/metabolismo , Lisossomos/enzimologia , Sinais Direcionadores de Proteínas , Proteínas Recombinantes/metabolismo , Sulfatases/metabolismo , Acetilglucosaminidase/genética , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Camundongos , Proteínas Recombinantes/genética , Sulfatases/genética
9.
Biochem J ; 478(17): 3185-3204, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34405853

RESUMO

p97 protein is a highly conserved, abundant, functionally diverse, structurally dynamic homohexameric AAA enzyme-containing N, D1, and D2 domains. A truncated p97 protein containing the N and D1 domains and the D1-D2 linker (ND1L) exhibits 79% of wild-type (WT) ATPase activity whereas the ND1 domain alone without the linker only has 2% of WT activity. To investigate the relationship between the D1-D2 linker and the D1 domain, we produced p97 ND1L mutants and demonstrated that this 22-residue linker region is essential for D1 ATPase activity. The conserved amino acid leucine 464 (L464) is critical for regulating D1 and D2 ATPase activity by p97 cofactors p37, p47, and Npl4-Ufd1 (NU). Changing leucine to alanine, proline, or glutamate increased the maximum rate of ATP turnover (kcat) of p47-regulated ATPase activities for these mutants, but not for WT. p37 and p47 increased the kcat of the proline substituted linker, suggesting that they induced linker conformations facilitating ATP hydrolysis. NU inhibited D1 ATPase activities of WT and mutant ND1L proteins, but activated D2 ATPase activity of full-length p97. To further understand the mutant mechanism, we used single-particle cryo-EM to visualize the full-length p97L464P and revealed the conformational change of the D1-D2 linker, resulting in a movement of the helix-turn-helix motif (543-569). Taken together with the biochemical and structural results we conclude that the linker helps maintain D1 in a competent conformation and relays the communication to/from the N-domain to the D1 and D2 ATPase domains, which are ∼50 Šaway.


Assuntos
Leucina/metabolismo , Domínios Proteicos/genética , Transdução de Sinais/genética , Proteína com Valosina/química , Proteína com Valosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Ativação Enzimática/genética , Células HeLa , Sequências Hélice-Volta-Hélice/genética , Humanos , Hidrólise , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação , Ligação Proteica/genética , Transfecção , Proteína com Valosina/genética
10.
Appl Environ Microbiol ; 87(15): e0020021, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-33990310

RESUMO

Characterizing the cell-level metabolic trade-offs that phytoplankton exhibit in response to changing environmental conditions is important for predicting the impact of these changes on marine food web dynamics and biogeochemical cycling. The time-selective proteome-labeling approach, bioorthogonal noncanonical amino acid tagging (BONCAT), has potential to provide insight into differential allocation of resources at the cellular level, especially when coupled with proteomics. However, the application of this technique in marine phytoplankton remains limited. We demonstrate that the marine cyanobacteria Synechococcus sp. and two groups of eukaryotic algae take up the modified amino acid l-homopropargylglycine (HPG), suggesting that BONCAT can be used to detect translationally active phytoplankton. However, the impact of HPG addition on growth dynamics varied between groups of phytoplankton. In addition, proteomic analysis of Synechococcus cells grown with HPG revealed a physiological shift in nitrogen metabolism, general protein stress, and energy production, indicating a potential limitation for the use of BONCAT in understanding the cell-level response of Synechococcus sp. to environmental change. Variability in HPG sensitivity between algal groups and the impact of HPG on Synechococcus physiology indicates that particular considerations should be taken when applying this technique to other marine taxa or mixed marine microbial communities. IMPORTANCE Phytoplankton form the base of the marine food web and substantially impact global energy and nutrient flow. Marine picocyanobacteria of the genus Synechococcus comprise a large portion of phytoplankton biomass in the ocean and therefore are important model organisms. The technical challenges of environmental proteomics in mixed microbial communities have limited our ability to detect the cell-level adaptations of phytoplankton communities to a changing environment. The proteome labeling technique, bioorthogonal noncanonical amino acid tagging (BONCAT), has potential to address some of these challenges by simplifying proteomic analyses. This study explores the ability of marine phytoplankton to take up the modified amino acid, l-homopropargylglycine (HPG), required for BONCAT, and investigates the proteomic response of Synechococcus to HPG. We not only demonstrate that cyanobacteria can take up HPG but also highlight the physiological impact of HPG on Synechococcus, which has implications for future applications of this technique in the marine environment.


Assuntos
Alcinos/farmacologia , Glicina/análogos & derivados , Fitoplâncton/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Synechococcus/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Glicina/farmacologia , Nitrogênio/metabolismo , Fitoplâncton/metabolismo , Proteoma/efeitos dos fármacos , Proteômica , Synechococcus/crescimento & desenvolvimento , Synechococcus/metabolismo
11.
Mol Pharm ; 18(1): 214-227, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33320673

RESUMO

There is currently no cure or effective treatment available for mucopolysaccharidosis type IIID (MPS IIID, Sanfilippo syndrome type D), a lysosomal storage disorder (LSD) caused by the deficiency of α-N-acetylglucosamine-6-sulfatase (GNS). The clinical symptoms of MPS IIID, like other subtypes of Sanfilippo syndrome, are largely localized to the central nervous system (CNS), and any treatments aiming to ameliorate or reverse the catastrophic and fatal neurologic decline caused by this disease need to be delivered across the blood-brain barrier. Here, we report a proof-of-concept enzyme replacement therapy (ERT) for MPS IIID using recombinant human α-N-acetylglucosamine-6-sulfatase (rhGNS) via intracerebroventricular (ICV) delivery in a neonatal MPS IIID mouse model. We overexpressed and purified rhGNS from CHO cells with a specific activity of 3.9 × 104 units/mg protein and a maximal enzymatic activity at lysosomal pH (pH 5.6), which was stable for over one month at 4 °C in artificial cerebrospinal fluid (CSF). We demonstrated that rhGNS was taken up by MPS IIID patient fibroblasts via the mannose 6-phosphate (M6P) receptor and reduced intracellular glycosaminoglycans to normal levels. The delivery of 5 µg of rhGNS into the lateral cerebral ventricle of neonatal MPS IIID mice resulted in normalization of the enzymatic activity in brain tissues; rhGNS was found to be enriched in lysosomes in MPS IIID-treated mice relative to the control. Furthermore, a single dose of rhGNS was able to reduce the accumulated heparan sulfate and ß-hexosaminidase. Our results demonstrate that rhGNS delivered into CSF is a potential therapeutic option for MPS IIID that is worthy of further development.


Assuntos
Mucopolissacaridose III/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Sulfatases/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Cricetulus , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doenças por Armazenamento dos Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Mucopolissacaridose III/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptor IGF Tipo 2/metabolismo
12.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34360842

RESUMO

IBMPFD/ALS is a genetic disorder caused by a single amino acid mutation on the p97 ATPase, promoting ATPase activity and cofactor dysregulation. The disease mechanism underlying p97 ATPase malfunction remains unclear. To understand how the mutation alters the ATPase regulation, we assembled a full-length p97R155H with its p47 cofactor and first visualized their structures using single-particle cryo-EM. More than one-third of the population was the dodecameric form. Nucleotide presence dissociates the dodecamer into two hexamers for its highly elevated function. The N-domains of the p97R155H mutant all show up configurations in ADP- or ATPγS-bound states. Our functional and structural analyses showed that the p47 binding is likely to impact the p97R155H ATPase activities via changing the conformations of arginine fingers. These functional and structural analyses underline the ATPase dysregulation with the miscommunication between the functional modules of the p97R155H.


Assuntos
Demência Frontotemporal/metabolismo , Modelos Moleculares , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mutação , Miosite de Corpos de Inclusão/metabolismo , Osteíte Deformante/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Proteína com Valosina/genética , Demência Frontotemporal/genética , Humanos , Microscopia Eletrônica de Transmissão , Distrofia Muscular do Cíngulo dos Membros/genética , Miosite de Corpos de Inclusão/genética , Osteíte Deformante/genética , Conformação Proteica , Proteína com Valosina/metabolismo
14.
Proc Natl Acad Sci U S A ; 112(14): E1705-14, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25775548

RESUMO

Dominant mutations in p97/VCP (valosin-containing protein) cause a rare multisystem degenerative disease with varied phenotypes that include inclusion body myopathy, Paget's disease of bone, frontotemporal dementia, and amyotrophic lateral sclerosis. p97 disease mutants have altered N-domain conformations, elevated ATPase activity, and altered cofactor association. We have now discovered a previously unidentified disease-relevant functional property of p97 by identifying how the cofactors p37 and p47 regulate p97 ATPase activity. We define p37 as, to our knowledge, the first known p97-activating cofactor, which enhances the catalytic efficiency (kcat/Km) of p97 by 11-fold. Whereas both p37 and p47 decrease the Km of ATP in p97, p37 increases the kcat of p97. In contrast, regulation by p47 is biphasic, with decreased kcat at low levels but increased kcat at higher levels. By deleting a region of p47 that lacks homology to p37 (amino acids 69-92), we changed p47 from an inhibitory cofactor to an activating cofactor, similar to p37. Our data suggest that cofactors regulate p97 ATPase activity by binding to the N domain. Induced conformation changes affect ADP/ATP binding at the D1 domain, which in turn controls ATPase cycling. Most importantly, we found that the D2 domain of disease mutants failed to be activated by p37 or p47. Our results show that cofactors play a critical role in controlling p97 ATPase activity, and suggest that lack of cofactor-regulated communication may contribute to p97-associated disease pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mutação , Trifosfato de Adenosina/metabolismo , Autofagia , Doenças Ósseas/metabolismo , Linhagem Celular Tumoral , Cromatografia em Gel , Complexo de Golgi , Homeostase , Humanos , Doenças Musculares/metabolismo , Doenças Neurodegenerativas/metabolismo , Fenótipo , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Proteína com Valosina
15.
Biochem Biophys Res Commun ; 491(3): 760-766, 2017 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-28739258

RESUMO

Human histidine triad nucleotide binding protein 1 (hHint1) is a purine nucleoside phosphoramidase and adenylate hydrolase that has emerged as a potential therapeutic target for the management of pain. However, the molecular mechanism of Hint1 in the signaling pathway has remained less clear. The role of metal ions in regulating postsynaptic transmission is well known, and the active site of hHint1 contains multiple histidines. Here we have investigated the effect of divalent metal ions (Cd2+, Cu2+, Mg2+, Mn2+, Ni2+, and Zn2+) on the structural integrity and catalytic activity of hHint1. With the exception of Mg2+, all the divalent ions inhibited hHint1, the rank of order was found to be Cu2+ >Zn2+ >Cd2+ ≥Ni2+ >Mn2+ based on their IC50 and kin/KI values. A crystal structure of hHint1 with bound Cu2+ is described to explain the competitive reversible inactivation of hHint1 by divalent cations. All the metal ions exhibited time- and concentration- dependent inhibition, with the rate of inactivation highly dependent on alterations of the C-terminus. With the exception of Cu2+; restoration of inhibition was observed for all the metal ions after treatment with EDTA. Our studies reveal a loss in secondary structure and aggregation of hHint1 upon incubation with 10-fold excess of copper. Thus, hHint1 appears to be structurally sensitive to irreversible inactivation by copper, which may be of neurotoxicological and pharmacological significance.


Assuntos
Metais/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/ultraestrutura , Analgésicos Opioides/química , Animais , Sítios de Ligação , Catálise , Cátions Bivalentes/química , Cobre/química , Humanos , Íons , Neuralgia/metabolismo , Ligação Proteica , Desdobramento de Proteína
16.
PLoS Pathog ; 11(5): e1004842, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25974051

RESUMO

Dialysis patients with chronic renal failure receiving deferoxamine for treating iron overload are uniquely predisposed for mucormycosis, which is most often caused by Rhizopus oryzae. Although the deferoxamine siderophore is not secreted by Mucorales, previous studies established that Rhizopus species utilize iron from ferrioxamine (iron-rich form of deferoxamine). Here we determined that the CBS domain proteins of Fob1 and Fob2 act as receptors on the cell surface of R. oryzae during iron uptake from ferrioxamine. Fob1 and Fob2 cell surface expression was induced in the presence of ferrioxamine and bound radiolabeled ferrioxamine. A R. oryzae strain with targeted reduced Fob1/Fob2 expression was impaired for iron uptake, germinating, and growing on medium with ferrioxamine as the sole source of iron. This strain also exhibited reduced virulence in a deferoxamine-treated, but not the diabetic ketoacidotic (DKA), mouse model of mucormycosis. The mechanism by which R. oryzae obtains iron from ferrioxamine involves the reductase/permease uptake system since the growth on ferrioxamine supplemented medium is associated with elevated reductase activity and the use of the ferrous chelator bathophenanthroline disulfonate abrogates iron uptake and growth on medium supplemented with ferrioxamine as a sole source of iron. Finally, R. oryzae mutants with reduced copies of the high affinity iron permease (FTR1) or with decreased FTR1 expression had an impaired iron uptake from ferrioxamine in vitro and reduced virulence in the deferoxamine-treated mouse model of mucormycosis. These two receptors appear to be conserved in Mucorales, and can be the subject of future novel therapy to maintain the use of deferoxamine for treating iron-overload.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Desferroxamina/metabolismo , Compostos Férricos/metabolismo , Ferro/metabolismo , Glicoproteínas de Membrana/metabolismo , Mucormicose/tratamento farmacológico , Rhizopus/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Virulência/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Quelantes de Ferro/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Mucormicose/microbiologia , Sideróforos/metabolismo
17.
Proc Natl Acad Sci U S A ; 111(37): E3880-9, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25187565

RESUMO

Many metabolic pathways are critically regulated during development and aging but little is known about the molecular mechanisms underlying this regulation. One key metabolic cascade in eukaryotes is the mevalonate pathway. It catalyzes the synthesis of sterol and nonsterol isoprenoids, such as cholesterol and ubiquinone, as well as other metabolites. In humans, an age-dependent decrease in ubiquinone levels and changes in cholesterol homeostasis suggest that mevalonate pathway activity changes with age. However, our knowledge of the mechanistic basis of these changes remains rudimentary. We have identified a regulatory circuit controlling the sumoylation state of Caenorhabditis elegans HMG-CoA synthase (HMGS-1). This protein is the ortholog of human HMGCS1 enzyme, which mediates the first committed step of the mevalonate pathway. In vivo, HMGS-1 undergoes an age-dependent sumoylation that is balanced by the activity of ULP-4 small ubiquitin-like modifier protease. ULP-4 exhibits an age-regulated expression pattern and a dynamic cytoplasm-to-mitochondria translocation. Thus, spatiotemporal ULP-4 activity controls the HMGS-1 sumoylation state in a mechanism that orchestrates mevalonate pathway activity with the age of the organism. To expand the HMGS-1 regulatory network, we combined proteomic analyses with knockout studies and found that the HMGS-1 level is also governed by the ubiquitin-proteasome pathway. We propose that these conserved molecular circuits have evolved to govern the level of mevalonate pathway flux during aging, a flux whose dysregulation is associated with numerous age-dependent cardiovascular and cancer pathologies.


Assuntos
Envelhecimento/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/fisiologia , Hidroximetilglutaril-CoA Sintase/fisiologia , Redes e Vias Metabólicas , Ácido Mevalônico/metabolismo , Sumoilação , Animais , Citosol/metabolismo , Humanos , Lisina/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Mutação/genética , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas , Transporte Proteico , Ubiquitina/metabolismo
18.
Brain ; 137(Pt 11): 2897-902, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25125609

RESUMO

Mutations in VCP have been reported to account for a spectrum of phenotypes that include inclusion body myopathy with Paget's disease of the bone and frontotemporal dementia, hereditary spastic paraplegia, and 1-2% of familial amyotrophic lateral sclerosis. We identified a novel VCP mutation (p.Glu185Lys) segregating in an autosomal dominant Charcot-Marie-Tooth disease type 2 family. Functional studies showed that the Glu185Lys variant impaired autophagic function leading to the accumulation of immature autophagosomes. VCP mutations should thus be considered for genetically undefined Charcot-Marie-Tooth disease type 2.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Ciclo Celular/genética , Doença de Charcot-Marie-Tooth/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Charcot-Marie-Tooth/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Linhagem , Fenótipo , Proteína com Valosina
19.
Proc Natl Acad Sci U S A ; 108(12): 4834-9, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21383145

RESUMO

A specific small-molecule inhibitor of p97 would provide an important tool to investigate diverse functions of this essential ATPase associated with diverse cellular activities (AAA) ATPase and to evaluate its potential to be a therapeutic target in human disease. We carried out a high-throughput screen to identify inhibitors of p97 ATPase activity. Dual-reporter cell lines that simultaneously express p97-dependent and p97-independent proteasome substrates were used to stratify inhibitors that emerged from the screen. N2,N4-dibenzylquinazoline-2,4-diamine (DBeQ) was identified as a selective, potent, reversible, and ATP-competitive p97 inhibitor. DBeQ blocks multiple processes that have been shown by RNAi to depend on p97, including degradation of ubiquitin fusion degradation and endoplasmic reticulum-associated degradation pathway reporters, as well as autophagosome maturation. DBeQ also potently inhibits cancer cell growth and is more rapid than a proteasome inhibitor at mobilizing the executioner caspases-3 and -7. Our results provide a rationale for targeting p97 in cancer therapy.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/metabolismo , Autofagia/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Inibidores Enzimáticos/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Quinazolinas/farmacologia , Ubiquitina/metabolismo , Adenosina Trifosfatases/genética , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 7/genética , Caspase 7/metabolismo , Linhagem Celular , Retículo Endoplasmático/genética , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Proteínas Nucleares/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Quinazolinas/síntese química , Quinazolinas/química , Ubiquitina/genética
20.
Int J Biol Macromol ; 254(Pt 2): 127905, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37939778

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a significant obstacle to lowering global cancer deaths. CB-5083, a novel valosin-containing protein (VCP)/p97 inhibitor that disrupts proteasomal degradation and induces endoplasmic reticulum stress (ERS) accumulation, was evaluated as an inducer of immunogenic cell death (ICD) in PDAC treatment. Furthermore, miR-142 enhances checkpoint blockade and promotes M1 repolarization, while Toll-like receptor 7/8 agonist resiquimod (R) acts as an immunoadjuvant to amplify the immune response to miR-142. This research signifies the first integration of CB, miR-142, and R in solid lipid nanoparticles (SLNs) modified with peptides targeting PD-L1, EGFR, and ER, which were shelled by the PEG-polyglutamic (PGA) coating that detaches in response to the acidic pH values in the tumor microenvironment (TME). The modified SLNs exhibited pH-sensitive cytotoxicity against Panc-02 cells, preserving normal cells and preventing hemolysis. The innovative approach simultaneously modulated pathways, including VCP/Bip/K48-Ub/ATF6, IRE1α/XBPs/LC3II, PD-L1/TGF-ß/IL-10/CD206/MSR1/Arg1, and TNF-α/IFN-γ/IL-6/iNOS/COX-2. Combined treatment blocked VCP, arrested the cell cycle, inhibited EMT, triggered ERS-mediated autophagy/apoptosis, and stimulated robust ICD via the release of damage-associated molecular patterns. This adaptable nanoformulation, displaying pH-sensitive PEG-PGA de-coating and precisely targeting EGFR, PD-L1, and ER, serves to hinder EMT and immune evasion, subsequently amplifying ICD in PDAC cells and the TME.


Assuntos
MicroRNAs , Neoplasias Pancreáticas , Humanos , Antígeno B7-H1 , Adjuvantes Imunológicos , Microambiente Tumoral , Endorribonucleases , Proteínas Serina-Treonina Quinases/metabolismo , Estresse do Retículo Endoplasmático , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Receptores ErbB , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA