Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 546
Filtrar
1.
PLoS One ; 19(8): e0307902, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39116118

RESUMO

Microcephaly, Guillain-Barré syndrome, and potential sexual transmission stand as prominent complications associated with Zika virus (ZIKV) infection. The absence of FDA-approved drugs or vaccines presents a substantial obstacle in combatting the virus. Furthermore, the inclusion of pregnancy in the pharmacological screening process complicates and extends the endeavor to ensure molecular safety and minimal toxicity. Given its pivotal role in viral assembly and maturation, the NS2B-NS3 viral protease emerges as a promising therapeutic target against ZIKV. In this context, a dipeptide inhibitor was specifically chosen as a control against 200 compounds for docking analysis. Subsequent molecular dynamics simulations extending over 200 ns were conducted to ascertain the stability of the docked complex and confirm the binding of the inhibitor at the protein's active site. The simulation outcomes exhibited conformity to acceptable thresholds, encompassing parameters such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), ligand-protein interaction analysis, ligand characterization, and surface area analysis. Notably, analysis of ligand angles bolstered the identification of prospective ligands capable of inhibiting viral protein activity and impeding virus dissemination. In this study, the integration of molecular docking and dynamics simulations has pinpointed the dipeptide inhibitor as a potential candidate ligand against ZIKV protease, thereby offering promise for therapeutic intervention against the virus.


Assuntos
Dipeptídeos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteases , Proteínas não Estruturais Virais , Zika virus , Zika virus/enzimologia , Zika virus/efeitos dos fármacos , Dipeptídeos/química , Dipeptídeos/farmacologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Antivirais/farmacologia , Antivirais/química , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Humanos , Ligação Proteica , Proteases Virais , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
2.
Nat Commun ; 15(1): 6080, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39030239

RESUMO

Dengue fever represents a significant medical and socio-economic burden in (sub)tropical regions, yet antivirals for treatment or prophylaxis are lacking. JNJ-A07 was described as highly active against the different genotypes within each serotype of the disease-causing dengue virus (DENV). Based on clustering of resistance mutations it has been assumed to target DENV non-structural protein 4B (NS4B). Using a photoaffinity labeling compound with high structural similarity to JNJ-A07, here we demonstrate binding to NS4B and its precursor NS4A-2K-NS4B. Consistently, we report recruitment of the compound to intracellular sites enriched for these proteins. We further specify the mechanism-of-action of JNJ-A07, which has virtually no effect on viral polyprotein cleavage, but targets the interaction between the NS2B/NS3 protease/helicase complex and the NS4A-2K-NS4B cleavage intermediate. This interaction is functionally linked to de novo formation of vesicle packets (VPs), the sites of DENV RNA replication. JNJ-A07 blocks VPs biogenesis with little effect on established ones. A similar mechanism-of-action was found for another NS4B inhibitor, NITD-688. In summary, we unravel the antiviral mechanism of these NS4B-targeting molecules and show how DENV employs a short-lived cleavage intermediate to carry out an early step of the viral life cycle.


Assuntos
Antivirais , Vírus da Dengue , Dengue , Proteínas não Estruturais Virais , Replicação Viral , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Humanos , Dengue/virologia , Dengue/tratamento farmacológico , Sorogrupo , RNA Helicases/metabolismo , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Ligação Proteica , Animais , Organelas/metabolismo , Organelas/efeitos dos fármacos , Proteases Virais , Aminofenóis , Proteínas de Membrana , Indóis , RNA Helicases DEAD-box , Nucleosídeo-Trifosfatase , Butiratos
3.
EMBO Mol Med ; 16(8): 1817-1839, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39009885

RESUMO

Zika virus (ZIKV) infection may lead to severe neurological consequences, including seizures, and early infancy death. However, the involved mechanisms are still largely unknown. TRPC channels play an important role in regulating nervous system excitability and are implicated in seizure development. We investigated whether TRPCs might be involved in the pathogenesis of ZIKV infection. We found that ZIKV infection increases TRPC4 expression in host cells via the interaction between the ZIKV-NS3 protein and CaMKII, enhancing TRPC4-mediated calcium influx. Pharmacological inhibition of CaMKII decreased both pCREB and TRPC4 protein levels, whereas the suppression of either TRPC4 or CaMKII improved the survival rate of ZIKV-infected cells and reduced viral protein production, likely by impeding the replication phase of the viral life cycle. TRPC4 or CaMKII inhibitors also reduced seizures and increased the survival of ZIKV-infected neonatal mice and blocked the spread of ZIKV in brain organoids derived from human-induced pluripotent stem cells. These findings suggest that targeting CaMKII or TRPC4 may offer a promising approach for developing novel anti-ZIKV therapies, capable of preventing ZIKV-associated seizures and death.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Canais de Cátion TRPC , Infecção por Zika virus , Zika virus , Infecção por Zika virus/virologia , Infecção por Zika virus/metabolismo , Animais , Humanos , Zika virus/fisiologia , Zika virus/efeitos dos fármacos , Camundongos , Canais de Cátion TRPC/metabolismo , Canais de Cátion TRPC/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Células HEK293 , Proteínas Virais/metabolismo , Convulsões/virologia , Convulsões/metabolismo , Convulsões/tratamento farmacológico , Proteases Virais , Serina Endopeptidases , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
4.
Antiviral Res ; 228: 105939, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38909960

RESUMO

Viruses have developed sophisticated strategies to control metabolic activity of infected cells in order to supply replication machinery with energy and metabolites. Dengue virus (DENV), a mosquito-borne flavivirus responsible for dengue fever, is no exception. Previous reports have documented DENV interactions with metabolic pathways and shown in particular that glycolysis is increased in DENV-infected cells. However, underlying molecular mechanisms are still poorly characterized and dependence of DENV on this pathway has not been investigated in details yet. Here, we identified an interaction between the non-structural protein 3 (NS3) of DENV and glucokinase regulator protein (GCKR), a host protein that inhibits the liver-specific hexokinase GCK. NS3 expression was found to increase glucose consumption and lactate secretion in hepatic cell line expressing GCK. Interestingly, we observed that GCKR interaction with GCK decreases DENV replication, indicating the dependence of DENV to GCK activity and supporting the role of NS3 as an inhibitor of GCKR function. Accordingly, in the same cells, DENV replication both induces and depends on glycolysis. By targeting NAD(H) biosynthesis with the antimetabolite 6-Amino-Nicotinamide (6-AN), we decreased cellular glycolytic activity and inhibited DENV replication in hepatic cells. Infection of primary organotypic liver cultures (OLiC) from hamsters was also inhibited by 6-AN. Altogether, our results show that DENV has evolved strategies to control glycolysis in the liver, which could account for hepatic dysfunctions associated to infection. Besides, our findings suggest that lowering intracellular availability of NAD(H) could be a valuable therapeutic strategy to control glycolysis and inhibit DENV replication in the liver.


Assuntos
Vírus da Dengue , Dengue , Glucoquinase , Glicólise , NAD , Proteínas não Estruturais Virais , Replicação Viral , Glicólise/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Glucoquinase/metabolismo , Glucoquinase/antagonistas & inibidores , Humanos , Replicação Viral/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Animais , Dengue/tratamento farmacológico , Dengue/virologia , Dengue/metabolismo , NAD/metabolismo , NAD/biossíntese , Linhagem Celular , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Glucose/metabolismo , Fígado/virologia , Fígado/metabolismo , Antivirais/farmacologia , Proteases Virais , Serina Endopeptidases , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
5.
Nucleic Acids Res ; 52(13): 7447-7464, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-38884215

RESUMO

The Orthoflavivirus NS3 helicase (NS3h) is crucial in virus replication, representing a potential drug target for pathogenesis. NS3h utilizes nucleotide triphosphate (ATP) for hydrolysis energy to translocate on single-stranded nucleic acids, which is an important step in the unwinding of double-stranded nucleic acids. Intermediate states along the ATP hydrolysis cycle and conformational changes between these states, represent important yet difficult-to-identify targets for potential inhibitors. Extensive molecular dynamics simulations of West Nile virus NS3h+ssRNA in the apo, ATP, ADP+Pi and ADP bound states were used to model the conformational ensembles along this cycle. Energetic and structural clustering analyses depict a clear trend of differential enthalpic affinity of NS3h with ADP, demonstrating a probable mechanism of hydrolysis turnover regulated by the motif-VI loop (MVIL). Based on these results, MVIL mutants (D471L, D471N and D471E) were found to have a substantial reduction in ATPase activity and RNA replication compared to the wild-type. Simulations of the mutants in the apo state indicate a shift in MVIL populations favoring either a closed or open 'valve' conformation, affecting ATP entry or stabilization, respectively. Combining our molecular modeling with experimental evidence highlights a conformation-dependent role for MVIL as a 'valve' for the ATP-pocket, presenting a promising target for antiviral development.


Assuntos
Trifosfato de Adenosina , Simulação de Dinâmica Molecular , RNA Helicases , Proteínas não Estruturais Virais , Vírus do Nilo Ocidental , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/genética , RNA Helicases/metabolismo , RNA Helicases/química , RNA Helicases/genética , Trifosfato de Adenosina/metabolismo , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/química , Motivos de Aminoácidos , Mutação , Nucleotídeos/metabolismo , Nucleotídeos/química , Hidrólise , Replicação Viral/genética , Conformação Proteica , Proteases Virais , Serina Endopeptidases , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
6.
SLAS Discov ; 29(5): 100164, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38796112

RESUMO

Zika virus (ZIKV) continues to pose a significant global public health threat, with recurring regional outbreaks and potential for pandemic spread. Despite often being asymptomatic, ZIKV infections can have severe consequences, including neurological disorders and congenital abnormalities. Unfortunately, there are currently no approved vaccines or antiviral drugs for the prevention or treatment of ZIKV. One promising target for drug development is the ZIKV NS2B-NS3 protease due to its crucial role in the virus life cycle. In this study, we established a cell-based ZIKV protease inhibition assay designed for high-throughput screening (HTS). Our assay relies on the ZIKV protease's ability to cleave a cyclised firefly luciferase fused to a natural cleavage sequence between NS2B and NS3 protease within living cells. We evaluated the performance of our assay in HTS setting using the pharmacologic controls (JNJ-40418677 and MK-591) and by screening a Library of Pharmacologically Active Compounds (LOPAC). The results confirmed the feasibility of our assay for compound library screening to identify potential ZIKV protease inhibitors.


Assuntos
Antivirais , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Inibidores de Proteases , Infecção por Zika virus , Zika virus , Zika virus/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Inibidores de Proteases/farmacologia , Humanos , Antivirais/farmacologia , Descoberta de Drogas/métodos , Infecção por Zika virus/virologia , Infecção por Zika virus/tratamento farmacológico , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Animais , Proteases Virais , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
7.
Antiviral Res ; 226: 105878, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582134

RESUMO

Flaviviruses can cause severe illness in humans. Effective and safe vaccines are available for some species; however, for many flaviviruses disease prevention or specific treatments remain unavailable. The viral replication cycle depends on the proteolytic activity of the NS2B-NS3 protease, which releases functional viral proteins from a non-functional polyprotein precursor, rendering the protease a promising drug target. In this study, we characterised recombinant NS2B-NS3 proteases from ten flaviviruses including three unreported proteases from the Usutu, Kyasanur forest disease and Powassan viruses. All protease constructs comprise a covalent Gly4-Ser-Gly4 linker connecting the NS3 serine protease domain with its cofactor NS2B. We conducted a comprehensive cleavage site analysis revealing areas of high conversion. While all proteases were active in enzymatic assays, we noted a 1000-fold difference in catalytic efficiency across proteases from different flaviviruses. Two bicyclic peptide inhibitors displayed anti-pan-flaviviral protease activity with inhibition constants ranging from 10 to 1000 nM.


Assuntos
Antivirais , Flavivirus , Serina Endopeptidases , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Flavivirus/efeitos dos fármacos , Flavivirus/enzimologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/química , Antivirais/farmacologia , Antivirais/química , Humanos , RNA Helicases/metabolismo , RNA Helicases/química , RNA Helicases/genética , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteases Virais , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
8.
Int J Biol Macromol ; 267(Pt 2): 131629, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38631585

RESUMO

Current management of HCV infection is based on Direct-Acting Antiviral Drugs (DAAs). However, resistance-associated mutations, especially in the NS3 and NS5B regions are gradually decreasing the efficacy of DAAs. Among the most effective HCV NS3/4A protease drugs, Sofosbuvir also develops resistance due to mutations in the NS3 and NS5B regions. Four mutations at positions A156Y, L36P, Q41H, and Q80K are classified as high-level resistance mutations. The resistance mechanism of HCV NS3/4A protease toward Sofosbuvir caused by these mutations is still unclear, as there is less information available regarding the structural and functional effects of the mutations against Sofosbuvir. In this work, we combined molecular dynamics simulation, molecular mechanics/Generalized-Born surface area calculation, principal component analysis, and free energy landscape analysis to explore the resistance mechanism of HCV NS3/4A protease due to these mutations, as well as compare interaction changes in wild-type. Subsequently, we identified that the mutant form of HCV NS3/4A protease affects the activity of Sofosbuvir. In this study, the resistance mechanism of Sofosbuvir at the atomic level is proposed. The proposed drug-resistance mechanism will provide valuable guidance for the design of HCV drugs.


Assuntos
Antivirais , Farmacorresistência Viral , Hepacivirus , Simulação de Dinâmica Molecular , Mutação , Sofosbuvir , Proteínas não Estruturais Virais , Antivirais/farmacologia , Antivirais/química , RNA Helicases DEAD-box , Farmacorresistência Viral/genética , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepacivirus/enzimologia , Nucleosídeo-Trifosfatase , Serina Endopeptidases , Serina Proteases , Sofosbuvir/farmacologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteases Virais
9.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38673962

RESUMO

In the global pandemic scenario, dengue and zika viruses (DENV and ZIKV, respectively), both mosquito-borne members of the flaviviridae family, represent a serious health problem, and considering the absence of specific antiviral drugs and available vaccines, there is a dire need to identify new targets to treat these types of viral infections. Within this drug discovery process, the protease NS2B/NS3 is considered the primary target for the development of novel anti-flavivirus drugs. The NS2B/NS3 is a serine protease that has a dual function both in the viral replication process and in the elusion of the innate immunity. To date, two main classes of NS2B/NS3 of DENV and ZIKV protease inhibitors have been discovered: those that bind to the orthosteric site and those that act at the allosteric site. Therefore, this perspective article aims to discuss the main features of the use of the most potent NS2B/NS3 inhibitors and their impact at the social level.


Assuntos
Antivirais , Dengue , Inibidores de Proteases , Infecção por Zika virus , Animais , Humanos , Antivirais/uso terapêutico , Antivirais/farmacologia , RNA Helicases DEAD-box , Dengue/tratamento farmacológico , Dengue/virologia , Vírus da Dengue/efeitos dos fármacos , Nucleosídeo-Trifosfatase , Inibidores de Proteases/uso terapêutico , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Serina Endopeptidases/metabolismo , Serina Endopeptidases/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Proteases Virais , Zika virus/efeitos dos fármacos , Zika virus/enzimologia , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/virologia
10.
J Basic Microbiol ; 64(6): e2300279, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38616711

RESUMO

Hepatitis C virus (HCV) is the most common infection worldwide. The correlation between HCV and renal cell carcinoma (RCC) is still mysterious. Therefore, the relationship between HCV and RCC was investigated. The study included 100 patients with RCC; 32 with HCV infection, and 68 without HCV infection. Expressions of viral proteins (NS3 and NS5A) were tested using an immune electron-microscope (IEM) and immunohistochemistry (IHC). IHC and quantitative real time-PCR investigated the presentation of human proteins TP53 and p21 genes. Transmission electron (TEM) detected viral-like particles in infected RCC tissues. The gene and protein expression of P53 was higher in HCV positive versus HCV negative patients and p21 was lower in HCV positive versus HCV negative in both tumor and normal tissue samples. Viral like particles were observed by TEM in the infected tumor and normal portion of the RCC tissues and the plasma samples. The IEM showed the depositions of NS3 and NS5A in infected renal tissues, while in noninfected samples, were not observed. The study hypothesizes that a correlation between HCV and RCC could exist through successfully detecting HCV-like particles, HCV proteins, and (p53 and p21) in RCC-infected patients.


Assuntos
Carcinoma de Células Renais , Genótipo , Hepacivirus , Neoplasias Renais , Proteína Supressora de Tumor p53 , Proteínas não Estruturais Virais , Humanos , Carcinoma de Células Renais/virologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Hepacivirus/genética , Proteínas não Estruturais Virais/genética , Neoplasias Renais/virologia , Neoplasias Renais/patologia , Neoplasias Renais/genética , Masculino , Proteína Supressora de Tumor p53/genética , Feminino , Pessoa de Meia-Idade , Hepatite C/virologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Idoso , Adulto , Imuno-Histoquímica , Proteases Virais , RNA Polimerase Dependente de RNA , RNA Helicases DEAD-box , Nucleosídeo-Trifosfatase , Serina Endopeptidases
11.
Protein Sci ; 33(4): e4934, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38501460

RESUMO

AlphaFold protein structure database (AlphaFold DB) archives a vast number of predicted models. We conducted systematic data mining against AlphaFold DB and discovered an uncharacterized P-loop NTPase family. The structure of the protein family was surprisingly novel, showing an atypical topology for P-loop NTPases, noticeable twofold symmetry, and two pairs of independent putative active sites. Our findings show that structural data mining is a powerful approach to identifying undiscovered protein families.


Assuntos
Nucleosídeo-Trifosfatase , Proteínas , Nucleosídeo-Trifosfatase/química , Nucleosídeo-Trifosfatase/metabolismo , Proteínas/química , Domínio Catalítico , Proteínas AAA/metabolismo
12.
J Biol Chem ; 300(3): 105711, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38309507

RESUMO

Cytosolic long dsRNA, among the most potent proinflammatory signals, is recognized by melanoma differentiation-associated protein 5 (MDA5). MDA5 binds dsRNA cooperatively forming helical filaments. ATP hydrolysis by MDA5 fulfills a proofreading function by promoting dissociation of shorter endogenous dsRNs from MDA5 while allowing longer viral dsRNAs to remain bound leading to activation of interferon-ß responses. Here, we show that adjacent MDA5 subunits in MDA5-dsRNA filaments hydrolyze ATP cooperatively, inducing cooperative filament disassembly. Consecutive rounds of ATP hydrolysis amplify the filament footprint, displacing tightly bound proteins from dsRNA. Our electron microscopy and biochemical assays show that LGP2 binds to dsRNA at internal binding sites through noncooperative ATP hydrolysis. Unlike MDA5, LGP2 has low nucleic acid selectivity and can hydrolyze GTP and CTP as well as ATP. Binding of LGP2 to dsRNA promotes nucleation of MDA5 filament assembly resulting in shorter filaments. Molecular modeling identifies an internally bound MDA5-LGP2-RNA complex, with the LGP2 C-terminal tail forming the key contacts with MDA5. These contacts are specifically required for NTP-dependent internal RNA binding. We conclude that NTPase-dependent binding of LGP2 to internal dsRNA sites complements NTPase-independent binding to dsRNA ends, via distinct binding modes, to increase the number and signaling output of MDA5-dsRNA complexes.


Assuntos
RNA Helicases DEAD-box , Helicase IFIH1 Induzida por Interferon , RNA Helicases , RNA de Cadeia Dupla , RNA Viral , Trifosfato de Adenosina/metabolismo , RNA Helicases DEAD-box/metabolismo , Hidrólise , Imunidade Inata , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Nucleosídeo-Trifosfatase/genética , Nucleosídeo-Trifosfatase/metabolismo , RNA Helicases/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Humanos
13.
Cell Rep ; 42(12): 113531, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38060382

RESUMO

Oocyte maturation is vital to attain full competence required for fertilization and embryogenesis. NLRP14 is preferentially expressed in mammalian oocytes and early embryos. Yet, the role and molecular mechanism of NLRP14 in oocyte maturation and early embryogenesis are poorly understood, and whether NLRP14 deficiency accounts for human infertility is unknown. Here, we found that maternal loss of Nlrp14 resulted in sterility with oocyte maturation defects and early embryonic arrest (EEA). Nlrp14 ablation compromised oocyte competence due to impaired cytoplasmic and nuclear maturation. Importantly, we revealed that NLRP14 maintained cytoplasmic UHRF1 abundance by protecting it from proteasome-dependent degradation and anchoring it from nuclear translocation in the oocyte. Furthermore, we identified compound heterozygous NLRP14 variants in women affected by infertility with EEA, which interrupted the NLRP14-UHRF1 interaction and decreased UHRF1 levels. Our data demonstrate NLRP14 as a cytoplasm-specific regulator of UHRF1 during oocyte maturation, providing insights into genetic diagnosis for female infertility.


Assuntos
Infertilidade Feminina , Animais , Feminino , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Oócitos/metabolismo , Oogênese , Citoplasma , Desenvolvimento Embrionário/genética , Mamíferos , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Nucleosídeo-Trifosfatase/metabolismo
14.
Adv Sci (Weinh) ; 10(27): e2301940, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37493331

RESUMO

Sperm-induced Ca2+ rise is critical for driving oocyte activation and subsequent embryonic development, but little is known about how lasting Ca2+ oscillations are regulated. Here it is shown that NLRP14, a maternal effect factor, is essential for keeping Ca2+ oscillations and early embryonic development. Few embryos lacking maternal NLRP14 can develop beyond the 2-cell stage. The impaired developmental potential of Nlrp14-deficient oocytes is mainly caused by disrupted cytoplasmic function and calcium homeostasis due to altered mitochondrial distribution, morphology, and activity since the calcium oscillations and development of Nlrp14-deficient oocytes can be rescued by substitution of whole cytoplasm by spindle transfer. Proteomics analysis reveal that cytoplasmic UHRF1 (ubiquitin-like, containing PHD and RING finger domains 1) is significantly decreased in Nlrp14-deficient oocytes, and Uhrf1-deficient oocytes also show disrupted calcium homeostasis and developmental arrest. Strikingly, it is found that the mitochondrial Na+ /Ca2+ exchanger (NCLX) encoded by Slc8b1 is significantly decreased in the Nlrp14mNull oocyte. Mechanistically, NLRP14 interacts with the NCLX intrinsically disordered regions (IDRs) domain and maintain its stability by regulating the K27-linked ubiquitination. Thus, the study reveals NLRP14 as a crucial player in calcium homeostasis that is important for early embryonic development.


Assuntos
Cálcio , Nucleosídeo-Trifosfatase , Sêmen , Humanos , Masculino , Cálcio/metabolismo , Homeostase/fisiologia , Oócitos/metabolismo , Sêmen/metabolismo , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo , Ubiquitinação , Animais , Camundongos , Nucleosídeo-Trifosfatase/metabolismo
15.
J Virol ; 97(7): e0051223, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37347173

RESUMO

Nonstructural protein 13 (nsp13), the helicase of SARS-CoV-2, has been shown to possess multiple functions that are essential for viral replication, and is considered an attractive target for the development of novel antivirals. We were initially interested in the interplay between nsp13 and interferon (IFN) signaling, and found that nsp13 inhibited reporter signal in an IFN-ß promoter assay. Surprisingly, the ectopic expression of different components of the RIG-I/MDA5 pathway, which were used to stimulate IFN-ß promoter, was also mitigated by nsp13. However, endogenous expression of these genes was not affected by nsp13. Interestingly, nsp13 restricted the expression of foreign genes originating from plasmid transfection, but failed to inhibit them after chromosome integration. These data, together with results from a runoff transcription assay and RNA sequencing, suggested a specific inhibition of episomal but not chromosomal gene transcription by nsp13. By using different truncated and mutant forms of nsp13, we demonstrated that its NTPase and helicase activities contributed to the inhibition of episomal DNA transcription, and that this restriction required direct interaction with episomal DNA. Based on these findings, we developed an economical and convenient high-throughput drug screening method targeting nsp13. We evaluated the inhibitory effects of various compounds on nsp13 by the expression of reporter gene plasmid after co-transfection with nsp13. In conclusion, we found that nsp13 can specifically inhibit episomal DNA transcription and developed a high-throughput drug screening method targeting nsp13 to facilitate the development of new antiviral drugs. IMPORTANCE To combat COVID-19, we need to understand SARS-CoV-2 and develop effective antiviral drugs. In our study, we serendipitously found that SARS-CoV-2 nsp13 could suppress episomal DNA transcription without affecting chromosomal DNA. Detailed characterization revealed that nsp13 suppresses episomal gene expression through its NTPase and helicase functions following DNA binding. Furthermore, we developed a high-throughput drug screening system targeting SARS-CoV-2 nsp13. Compared to traditional SARS-CoV-2 drug screening methods, our system is more economical and convenient, facilitating the development of more potent and selective nsp13 inhibitors and enabling the discovery of new antiviral therapies.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Nucleosídeo-Trifosfatase/genética , RNA Helicases/metabolismo , Proteínas não Estruturais Virais/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Antivirais/farmacologia , DNA , Plasmídeos/genética
16.
Plasmid ; 127: 102694, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37301314

RESUMO

Plasmid families harbor different maintenances functions, depending on their size and copy number. Low copy number plasmids rely on active partition systems, organizing a partition complex at specific centromere sites that is actively positioned using NTPase proteins. Some low copy number plasmids lack an active partition system, but carry atypical intracellular positioning systems using a single protein that binds to the centromere site but without an associated NTPase. These systems have been studied in the case of the Escherichia coli R388 and of the Staphylococcus aureus pSK1 plasmids. Here we review these two systems, which appear to be unrelated but share common features, such as their distribution on plasmids of medium size and copy number, certain activities of their centromere-binding proteins, StbA and Par, respectively, as well as their mode of action, which may involve dynamic interactions with the nucleoid-packed chromosome of their hosts.


Assuntos
Variações do Número de Cópias de DNA , Nucleosídeo-Trifosfatase , Humanos , Plasmídeos/genética , Nucleosídeo-Trifosfatase/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Bactérias/genética , Segregação de Cromossomos
17.
Nat Commun ; 14(1): 1886, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37019967

RESUMO

Activating signal co-integrator 1 complex (ASCC) subunit 3 (ASCC3) supports diverse genome maintenance and gene expression processes, and contains tandem Ski2-like NTPase/helicase cassettes crucial for these functions. Presently, the molecular mechanisms underlying ASCC3 helicase activity and regulation remain unresolved. We present cryogenic electron microscopy, DNA-protein cross-linking/mass spectrometry as well as in vitro and cellular functional analyses of the ASCC3-TRIP4 sub-module of ASCC. Unlike the related spliceosomal SNRNP200 RNA helicase, ASCC3 can thread substrates through both helicase cassettes. TRIP4 docks on ASCC3 via a zinc finger domain and stimulates the helicase by positioning an ASC-1 homology domain next to the C-terminal helicase cassette of ASCC3, likely supporting substrate engagement and assisting the DNA exit. TRIP4 binds ASCC3 mutually exclusively with the DNA/RNA dealkylase, ALKBH3, directing ASCC3 for specific processes. Our findings define ASCC3-TRIP4 as a tunable motor module of ASCC that encompasses two cooperating NTPase/helicase units functionally expanded by TRIP4.


Assuntos
DNA Helicases , Nucleosídeo-Trifosfatase , Nucleosídeo-Trifosfatase/metabolismo , DNA Helicases/metabolismo , Spliceossomos/metabolismo , RNA Helicases/metabolismo , DNA/metabolismo
18.
Nature ; 616(7955): 152-158, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36991121

RESUMO

Non-enveloped viruses require cell lysis to release new virions from infected cells, suggesting that these viruses require mechanisms to induce cell death. Noroviruses are one such group of viruses, but there is no known mechanism that causes norovirus infection-triggered cell death and lysis1-3. Here we identify a molecular mechanism of norovirus-induced cell death. We found that the norovirus-encoded NTPase NS3 contains an N-terminal four-helix bundle domain homologous to the membrane-disruption domain of the pseudokinase mixed lineage kinase domain-like (MLKL). NS3 has a mitochondrial localization signal and thus induces cell death by targeting mitochondria. Full-length NS3 and an N-terminal fragment of the protein bound the mitochondrial membrane lipid cardiolipin, permeabilized the mitochondrial membrane and induced mitochondrial dysfunction. Both the N-terminal region and the mitochondrial localization motif of NS3 were essential for cell death, viral egress from cells and viral replication in mice. These findings suggest that noroviruses have acquired a host MLKL-like pore-forming domain to facilitate viral egress by inducing mitochondrial dysfunction.


Assuntos
Morte Celular , Norovirus , Nucleosídeo-Trifosfatase , Proteínas Quinases , Proteínas Virais , Animais , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Norovirus/enzimologia , Norovirus/crescimento & desenvolvimento , Norovirus/patogenicidade , Norovirus/fisiologia , Proteínas Quinases/química , Replicação Viral , Proteínas Virais/química , Proteínas Virais/metabolismo , Nucleosídeo-Trifosfatase/química , Nucleosídeo-Trifosfatase/metabolismo , Sinais Direcionadores de Proteínas , Cardiolipinas/metabolismo , Membranas Mitocondriais/química , Membranas Mitocondriais/metabolismo
19.
Viruses ; 15(3)2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36992520

RESUMO

Human noroviruses (HuNVs) are the leading cause of gastroenteritis worldwide. NS1.2 is critical for HuNV pathogenesis, but the function is still unclear. The GII NS1.2 of HuNVs, unlike GI NS1.2, was localized to the endoplasmic reticulum (ER) and lipid droplets (LDs) and is accompanied by a distorted-filamentous ER morphology and aggregated-enlarged LDs. LC3 was recruited to the NS1.2-localized membrane through an autophagy-independent pathway. NS1.2, expressed from a cDNA clone of GII.4 norovirus, formed complexes with NTPase and NS4, which exhibited aggregated vesicle-like structures that were also colocalized with LC3 and LDs. NS1.2 is structurally divided into three domains from the N terminus: an inherently disordered region (IDR), a region that contains a putative hydrolase with the H-box/NC catalytic center (H-box/NC), and a C-terminal 251-330 a.a. region containing membrane-targeting domain. All three functional domains of NS1.2 were required for the induction of the filamentous ER. The IDR was essential for LC3 recruitment by NS1.2. Both the H-Box/NC and membrane-targeting domains are required for the induction of aggregated-enlarged LDs, NS1.2 self-assembly, and interaction with NTPase. The membrane-targeting domain was sufficient to interact with NS4. The study characterized the NS1.2 domain required for membrane targeting and protein-protein interactions, which are crucial for forming a viral replication complex.


Assuntos
Norovirus , Humanos , Norovirus/genética , Nucleosídeo-Trifosfatase , Gotículas Lipídicas/metabolismo , Replicação Viral/genética , Proteínas não Estruturais Virais/metabolismo , Retículo Endoplasmático/metabolismo
20.
PLoS Pathog ; 19(2): e1011179, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36848386

RESUMO

Chikungunya virus (CHIKV) is a reemerging alphavirus. Since 2005, it has infected millions of people during outbreaks in Africa, Asia, and South/Central America. CHIKV replication depends on host cell factors at many levels and is expected to have a profound effect on cellular physiology. To obtain more insight into host responses to infection, stable isotope labeling with amino acids in cell culture and liquid chromatography-tandem mass spectrometry were used to assess temporal changes in the cellular phosphoproteome during CHIKV infection. Among the ~3,000 unique phosphorylation sites analyzed, the largest change in phosphorylation status was measured on residue T56 of eukaryotic elongation factor 2 (eEF2), which showed a >50-fold increase at 8 and 12 h p.i. Infection with other alphaviruses (Semliki Forest, Sindbis and Venezuelan equine encephalitis virus (VEEV)) triggered a similarly strong eEF2 phosphorylation. Expression of a truncated form of CHIKV or VEEV nsP2, containing only the N-terminal and NTPase/helicase domains (nsP2-NTD-Hel), sufficed to induce eEF2 phosphorylation, which could be prevented by mutating key residues in the Walker A and B motifs of the NTPase domain. Alphavirus infection or expression of nsP2-NTD-Hel resulted in decreased cellular ATP levels and increased cAMP levels. This did not occur when catalytically inactive NTPase mutants were expressed. The wild-type nsP2-NTD-Hel inhibited cellular translation independent of the C-terminal nsP2 domain, which was previously implicated in directing the virus-induced host shut-off for Old World alphaviruses. We hypothesize that the alphavirus NTPase activates a cellular adenylyl cyclase resulting in increased cAMP levels, thus activating PKA and subsequently eukaryotic elongation factor 2 kinase. This in turn triggers eEF2 phosphorylation and translational inhibition. We conclude that the nsP2-driven increase of cAMP levels contributes to the alphavirus-induced shut-off of cellular protein synthesis that is shared between Old and New World alphaviruses. MS Data are available via ProteomeXchange with identifier PXD009381.


Assuntos
Alphavirus , Febre de Chikungunya , Vírus Chikungunya , Humanos , Alphavirus/metabolismo , Nucleosídeo-Trifosfatase/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Eucariotos , Fosforilação , Vírus Chikungunya/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Quinase do Fator 2 de Elongação/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...