Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Angew Chem Int Ed Engl ; 62(41): e202309657, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37609788

ABSTRACT

The main protease (Mpro ) of SARS-CoV-2 is a well-characterized target for antiviral drug discovery. To date, most antiviral drug discovery efforts have focused on the S4-S1' pocket of Mpro ; however, it is still unclear whether the S1'-S3' pocket per se can serve as a new site for drug discovery. In this study, the S1'-S3' pocket of Mpro was found to differentially recognize viral peptidyl substrates. For instance, S3' in Mpro strongly favors Phe or Trp, and S1' favors Ala. The peptidyl inhibitor D-4-77, which possesses an α-bromoacetamide warhead, was discovered to be a promising inhibitor of Mpro , with an IC50 of 0.95 µM and an antiviral EC50 of 0.49 µM. The Mpro /inhibitor co-crystal structure confirmed the binding mode of the inhibitor to the S1'-S3' pocket and revealed a covalent mechanism. In addition, D-4-77 functions as an immune protectant and suppresses SARS-CoV-2 Mpro -induced antagonism of the host NF-κB innate immune response. These findings indicate that the S1'-S3' pocket of SARS-CoV-2 Mpro is druggable, and that inhibiting SARS-CoV-2 Mpro can simultaneously protect human innate immunity and inhibit virion assembly.

2.
Transplant Cell Ther ; 29(7): 456.e1-456.e11, 2023 07.
Article in English | MEDLINE | ID: mdl-37028555

ABSTRACT

FLT3-ITD mutation has consistently been correlated with poor outcomes in patients with acute myeloid leukemia (AML). Allogeneic hematopoietic stem cell transplantation (allo-HSCT) plays a major role in curing blood diseases. Whether allo-HSCT can eliminate the detrimental effects of FLT3-ITD mutation in AML patients remains debatable. In addition, studies have shown that the FLT3-ITD allelic ratio (AR) and NPM1 mutation appear to further influence the prognostic utility of FLT3-ITD in patients with FLT3-ITD-mutated AML. The influence of NPM1 mutation and AR on FLT3-ITDmut patients in our database remains unclear. We aimed to compare survival outcomes following allo-HSCT between patients with FLT3-ITDmut and those with wild-type FLT3-ITD and to further analyze the influence of NPM1 and AR on outcomes. A total of 118 FLT3-ITDmut patients and 497 FLT3-ITDwt patients who underwent allo-HSCT were propensity score-matched 1:3 using nearest-neighbor matching with a caliper size of .2. The study cohort comprised 430 patients with AML, including 116 with FLT3-ITDmut and 314 with FLT3-ITDwt. Overall survival (OS) and leukemia-free survival (LFS) were similar in the FLT3-ITDmut patients and the FLT3-ITDwt patients (2-year OS,78.5% versus 82.6% [P = .374]; 2-year LFS, 75.1% versus 80.8% [P = .215]). A cutoff of .50 was applied to define subgroups with low and high FLT3-ITD AR. No significant differences in the cumulative incidence of relapse (CIR) or LFS were observed between the low AR and high AR groups (2-year CIR, P = .617; 2-year LFS, P = .563). CIR and LFS also were comparable when patients were grouped according to the presence or absence of NPM1 and FLT3-ITD (2-year CIR, P = .356; 2-year LFS, P = .159). Additionally, the CIR and LFS of FLT3-ITDmut and FLT3-ITDwt patients tended to differ after matched sibling donor HSCT (2-year CIR, P = .072; 2-year LFS, P = .084); however, these differences were not seen in recipients of haploidentical (haplo-) HSCT (2-year CIR, P = .59; 2-year LFS, P = .794). The presence of minimal residual disease before transplantation and lack of first complete response were identified as risk factors related to inferior outcomes in a multivariate analysis regardless of FLT3-ITD or NPM1 status. Our results suggest that allo-HSCT, especially haplo-HSCT, may overcome the adverse effect of FLT3-ITD mutation irrespective of NPM1 status or AR. Allo-HSCT could be an ideal option for AML patients with FLT3-ITD.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Humans , Retrospective Studies , Propensity Score , Nucleophosmin , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Recurrence , Mutation , fms-Like Tyrosine Kinase 3/genetics
3.
Int J Cancer ; 152(5): 977-985, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36251447

ABSTRACT

Previous studies have demonstrated that TP53 mutation is correlated with insufficient therapy response and unfavorable prognosis in acute lymphoblastic leukemia (ALL). Few studies have investigated the impact of TP53 mutation in ALL patients after haploidentical hematopoietic stem cell transplantation (haplo-HSCT). We completed a retrospective study of 65 ALL patients with available TP53 status who underwent haplo-HSCT. They were divided into a TP53 mutation group (TP53mut ) and a TP53 wild-type (TP53wt ) group. TP53mut showed comparable 2-year cumulative incidence of relapse (CIR) rates (13.1% vs 12.5%, P = .96) and 2-year leukemia-free survival (LFS) (74.2% vs 77.4%, P = .80) with TP53wt . No significant differences in 2-year overall survival (OS) rates (82.9% vs 87.3%, P = .61) or 2-year NRM rates (12.7% vs 10.2%, P = .69) were observed in TP53mut and TP53wt patients. Multivariate analysis suggested that white blood cell (WBC) count at initial diagnosis (>50 × 109 /L: hazard ratio [HR] = 3.860, P = .016) and age (>40 years old: HR = 4.120, P = .012) are independent risk factors for 2-year LFS. Our study showed that TP53 mutations may not be related to the unfavorable impact on survival in ALL patients after treatment with haplo-HSCT. The present results suggested that haplo-HSCT may eliminate the poor prognosis effect of TP53 mutation in ALL.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Tumor Suppressor Protein p53 , Adult , Humans , Genes, p53 , Graft vs Host Disease/etiology , Leukemia, Myeloid, Acute/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Retrospective Studies , Tumor Suppressor Protein p53/genetics
4.
Front Mol Biosci ; 8: 655687, 2021.
Article in English | MEDLINE | ID: mdl-34381810

ABSTRACT

Neuroinflammation is a key pathological factor in numerous neurological disorders. Cumulating evidence has indicated critical roles of NAD+/NADH metabolism in multiple major diseases, while the role of malate-aspartate shuttle (MAS) - a major NADH shuttle - in inflammation has remained unclear. In this study we investigated the roles of MAS in LPS-induced neuroinflammation both in vivo and in vitro. Immunofluorescence staining, Western blot assay and Real-time PCR assays were conducted to determine the activation of Iba-1, the protein levels of iNOS and COX2 and the mRNA levels of IL-1ß, IL-6, and TNF-α in vivo, showing that both pre-treatment and post-treatment of aminooxyacetic acid (AOAA) - an MAS inhibitor - profoundly decreased the LPS-induced neuroinflammation in mice. BV2 microglia was also used as a cellular model to investigate the mechanisms of this finding, in which such assays as Western blot assay and nitrite assay. Our study further indicated that AOAA produced its effects on LPS-induced microglial activation by its effects on MAS: Pyruvate treatment reversed the effects of AOAA on the cytosolic NAD+/NADH ratio, which also restored the LPS-induced activation of the AOAA-treated microglia. Moreover, the lactate dehydrogenase (LDH) inhibitor GSK2837808A blocked the effects of pyruvate on the AOAA-produced decreases in both the cytosolic NAD+/NADH ratio and LPS-induced microglial activation. Our study has further suggested that AOAA produced inhibition of LPS-induced microglial activation at least partially by decreasing STAT3 phosphorylation. Collectively, our findings have indicated AOAA as a new and effective drug for inhibiting LPS-induced neuroinflammation. Our study has also indicated that MAS is a novel mediator of LPS-induced neuroinflammation due to its capacity to modulate LPS-induced STAT3 phosphorylation, which has further highlighted a critical role of NAD+/NADH metabolism in inflammation.

5.
PeerJ ; 9: e11401, 2021.
Article in English | MEDLINE | ID: mdl-34040894

ABSTRACT

BACKGROUND: Multiple studies have indicated crucial roles of NAD+ deficiency in several neurological diseases and aging. It is critical to discover the mechanisms underlying the NAD+ deficiency. A decreased level of Nicotinamide phosphoribosyltransferase (Nampt)-an important enzyme in the salvage pathway of NAD+ synthesis-has been found under certain pathological conditions, while the mechanisms underlying the Nampt decrease are unclear. The purpose of this study is to test the hypothesis that oxidative stress can produce decreased Nampt, and to investigate the biological effects of Nampt on NAD+ synthesis and cell survival under both basal and oxidative stress conditions. METHODS: We used differentiated PC12 cells as a cellular model to investigate the effects of oxidative stress on the levels of Nampt. Multiple assays, including flow cytometry-based cell death assays and NAD+ assays were conducted. RESULTS: First, oxidative stress can decrease the levels of Nampt mRNA and Nampt protein; second, Nampt plays significant roles in NAD+ synthesis under both basal conditions and oxidative stress conditions; third, Nampt plays critical roles in cell survival under both basal conditions and oxidative stress conditions; and fourth, oxidative stress produced decreased NAD+ levels and cell survival partially by decreasing Nampt. Collectively, our study has indicated that oxidative stress is a pathological factor leading to decreased Nampt, which plays important roles in oxidative stress-produced decreases in NAD+ levels and cell survival. Our findings have indicated major roles of Nampt in maintaining NAD+ levels and cell survival under both basal and oxidative stress conditions.

6.
Proc Natl Acad Sci U S A ; 117(18): 9876-9883, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32303654

ABSTRACT

A massive intronic hexanucleotide repeat (GGGGCC) expansion in C9ORF72 is a genetic origin of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recently, C9ORF72, together with SMCR8 and WDR41, has been shown to regulate autophagy and function as Rab GEF. However, the precise function of C9ORF72 remains unclear. Here, we report the cryogenic electron microscopy (cryo-EM) structure of the human C9ORF72-SMCR8-WDR41 complex at a resolution of 3.2 Å. The structure reveals the dimeric assembly of a heterotrimer of C9ORF72-SMCR8-WDR41. Notably, the C-terminal tail of C9ORF72 and the DENN domain of SMCR8 play critical roles in the dimerization of the two protomers of the C9ORF72-SMCR8-WDR41 complex. In the protomer, C9ORF72 and WDR41 are joined by SMCR8 without direct interaction. WDR41 binds to the DENN domain of SMCR8 by the C-terminal helix. Interestingly, the prominent structural feature of C9ORF72-SMCR8 resembles that of the FLNC-FNIP2 complex, the GTPase activating protein (GAP) of RagC/D. Structural comparison and sequence alignment revealed that Arg147 of SMCR8 is conserved and corresponds to the arginine finger of FLCN, and biochemical analysis indicated that the Arg147 of SMCR8 is critical to the stimulatory effect of the C9ORF72-SMCR8 complex on Rab8a and Rab11a. Our study not only illustrates the basis of C9ORF72-SMCR8-WDR41 complex assembly but also reveals the GAP activity of the C9ORF72-SMCR8 complex.


Subject(s)
Autophagy-Related Proteins/ultrastructure , C9orf72 Protein/ultrastructure , Carrier Proteins/ultrastructure , Multiprotein Complexes/ultrastructure , Amino Acid Sequence/genetics , Amyotrophic Lateral Sclerosis/genetics , Arginine/genetics , Autophagy/genetics , Autophagy-Related Proteins/genetics , C9orf72 Protein/genetics , Carrier Proteins/genetics , Cryoelectron Microscopy , Filamins/genetics , Filamins/ultrastructure , Frontotemporal Dementia/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/ultrastructure , Genetic Predisposition to Disease , Humans , Multiprotein Complexes/genetics , Sequence Alignment , rab GTP-Binding Proteins/genetics
7.
Cell Discov ; 6: 7, 2020.
Article in English | MEDLINE | ID: mdl-32128251

ABSTRACT

Super elongation complex (SEC) is a positive regulator of RNA polymerase II, which is required for HIV-1 proviral transcription. AFF1/4 is the scaffold protein that recruits other components of SEC and forms dimer depending on its THD domain (TPRL with Handle Region Dimerization Domain). Here we report the crystal structure of the human AFF4-THD at the resolution of 2.4 Å. The α4, α5, and α6 of one AFF4-THD mediate the formation of a dimer and pack tightly against the equivalent part of the second molecule in the dimer of AFF-THD. Mutagenesis analysis revealed that single mutations of either Phe1014 or Tyr1096 of AFF4 to alanine impair the formation of the AFF4 dimer. In addition, transactivation assay also indicated that Phe1014 and Tyr1096 of AFF4 are critical to the transactivation activity of AFF4. Interestingly, the corresponding residues Phe1063 and Tyr1145 in AFF1 have an effect on the transactivation of HIV-1 provirus. However, such mutations of AFF1/4 have no effect on the interaction of AFF1/4 with other subunits of the SEC. Together, our data demonstrated that the dimerization of AFF1/4 is essential to transactivation of HIV-1 provirus.

8.
Nucleic Acids Res ; 44(11): 5231-45, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27001510

ABSTRACT

DNA damage tolerance (DDT) is responsible for genomic stability and cell viability by bypassing the replication block. In Saccharomyces cerevisiae DDT employs two parallel branch pathways to bypass the DNA lesion, namely translesion DNA synthesis (TLS) and error-free lesion bypass, which are mediated by sequential modifications of PCNA. Rad5 has been placed in the error-free branch of DDT because it contains an E3 ligase domain required for PCNA polyubiquitination. Rad5 is a multi-functional protein and may also play a role in TLS, since it interacts with the TLS polymerase Rev1. In this study we mapped the Rev1-interaction domain in Rad5 to the amino acid resolution and demonstrated that Rad5 is indeed involved in TLS possibly through recruitment of Rev1. Genetic analyses show that the dual functions of Rad5 can be separated and reconstituted. Crystal structure analysis of the Rad5-Rev1 interaction reveals a consensus RFF motif in the Rad5 N-terminus that binds to a hydrophobic pocket within the C-terminal domain of Rev1 that is highly conserved in eukaryotes. This study indicates that Rad5 plays a critical role in pathway choice between TLS and error-free DDT.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Nucleotidyltransferases/metabolism , Saccharomycetales/genetics , Saccharomycetales/metabolism , Amino Acid Sequence , DNA Damage , DNA Helicases/chemistry , Epistasis, Genetic , Models, Molecular , Mutation , Nucleotidyltransferases/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...