Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 9608, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38671207

RESUMO

Mountain tunnels portal often have to pass through slope terrain unavoidably, thus forming a shallow buried bias tunnel. During the construction of shallow buried bias tunnel, disasters such as slope sliding and tunnel collapse frequently occur. The failure mode of surrounding rock obtained by current research is based on the limit equilibrium theory, which cannot reflect the progressive failure characteristics of the surrounding rock of shallow buried bias tunnel. In order to reveal the failure mechanism of the gradual instability of surrounding rock of shallow buried bias tunnel, the problem of gradual failure of the surrounding rock is reduced to an elastic-plastic analysis problem for surrounding rock considering the strain-softening characteristics. Based on the elastic-plastic analysis of the failure process of shallow buried bias tunnel, MATLAB was used to compile a program to read the finite-difference calculation result file, extract the effective information such as shear strain and tensile strain at the center point of each unit, and establish the analysis method of the progressive failure mode of shallow buried bias tunnel. The reliability of the method proposed was verified by comparing the failure process of the model test with the development process of shear strain increment. Under the condition of no support, the formation mechanism of failure plane of surrounding rock on both sides of shallow buried bias tunnel is different. The shallow buried side is the shear failure plane formed by the collapse of surrounding rock, while the deep buried side of the tunnel is the shear failure plane formed by the collapse of surrounding rock and slope sliding. Under the conditions of excavation and support, the failure plane of the shallow buried bias tunnel can be divided into three parts according to the formation sequence and reasons. The part I is the failure plane, which is formed by active shear under the influence of tunnel excavation. The part II is the failure plane formed by tensile crack of slope top. The part III is the failure plane formed by passive shear under the push of the soil in the upper part of the slope.

2.
Int J Biol Macromol ; 264(Pt 1): 130542, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432272

RESUMO

Pathological cardiac hypertrophy (CH) is driven by maladaptive changes in myocardial cells in response to pressure overload or other stimuli. CH has been identified as a significant risk factor for the development of various cardiovascular diseases, ultimately resulting in heart failure. Melanoma differentiation-associated protein 5 (MDA5), encoded by interferon-induced with helicase C domain 1 (IFIH1), is a cytoplasmic sensor that primarily functions as a detector of double-stranded ribonucleic acid (dsRNA) viruses in innate immune responses; however, its role in CH pathogenesis remains unclear. Thus, the aim of this study was to examine the relationship between MDA5 and CH using cellular and animal models generated by stimulating neonatal rat cardiomyocytes with phenylephrine and by performing transverse aortic constriction on mice, respectively. MDA5 expression was upregulated in all models. MDA5 deficiency exacerbated myocardial pachynsis, fibrosis, and inflammation in vivo, whereas its overexpression hindered CH development in vitro. In terms of the underlying molecular mechanism, MDA5 inhibited CH development by promoting apoptosis signal-regulating kinase 1 (ASK1) phosphorylation, thereby suppressing c-Jun N-terminal kinase/p38 signaling pathway activation. Rescue experiments using an ASK1 activation inhibitor confirmed that ASK1 phosphorylation was essential for MDA5-mediated cell death. Thus, MDA5 protects against CH and is a potential therapeutic target.


Assuntos
Apoptose , MAP Quinase Quinase Quinase 5 , Camundongos , Ratos , Animais , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Apoptose/fisiologia , Cardiomegalia/metabolismo , Transdução de Sinais , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo
3.
Cell Death Discov ; 10(1): 142, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38490981

RESUMO

Pleckstrin homology domain-containing family M member 2 (PLEKHM2) is an essential adaptor for lysosomal trafficking and its homozygous truncation have been reported to cause early onset dilated cardiomyopathy (DCM). However, the molecular mechanism of PLEKHM2 deficiency in DCM pathogenesis and progression is poorly understood. Here, we generated an in vitro model of PLEKHM2 knockout (KO) induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to elucidate the potential pathogenic mechanism of PLEKHM2-deficient cardiomyopathy. PLEKHM2-KO hiPSC-CMs developed disease phenotypes with reduced contractility and impaired calcium handling. Subsequent RNA sequencing (RNA-seq) analysis revealed altered expression of genes involved in mitochondrial function, autophagy and apoptosis in PLEKHM2-KO hiPSC-CMs. Further molecular experiments confirmed PLEKHM2 deficiency impaired autophagy and resulted in accumulation of damaged mitochondria, which triggered increased reactive oxygen species (ROS) levels and decreased mitochondrial membrane potential (Δψm). Importantly, the elevated ROS levels caused oxidative stress-induced damage to nearby healthy mitochondria, resulting in extensive Δψm destabilization, and ultimately leading to impaired mitochondrial function and myocardial contractility. Moreover, ROS inhibition attenuated oxidative stress-induced mitochondrial damage, thereby partially rescued PLEKHM2 deficiency-induced disease phenotypes. Remarkably, PLEKHM2-WT overexpression restored autophagic flux and rescued mitochondrial function and myocardial contractility in PLEKHM2-KO hiPSC-CMs. Taken together, these results suggested that impaired mitochondrial clearance and increased ROS levels play important roles in PLEKHM2-deficient cardiomyopathy, and PLEKHM2-WT overexpression can improve mitochondrial function and rescue PLEKHM2-deficient cardiomyopathy.

4.
Stem Cell Res ; 73: 103225, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37931537

RESUMO

Toll-like receptor 4 (TLR4) is a pattern recognition receptor (PRRS) and an important protective immune receptor. TLR4 deficiency can lead to Inflammatory bowel disease. To explore the role of TLR4, we used CRISPR/Cas9 system to produce TLR4 compound heterozygous knockout embryonic stem cells in H9 cell line. The WAe009-A-N has a compound heterozygous 7 bp deletion/8 bp deletion in TLR4 exon 3, which resulted in a frameshift in the translation of TLR4, and TLR4 protein wasn't detectable in this cell line. In addition, the WAe009-A-N with normal karyotype can express pluripotent markers and differentiate into three germ layers in vitro.


Assuntos
Células-Tronco Embrionárias Humanas , Humanos , Células-Tronco Embrionárias Humanas/metabolismo , Sistemas CRISPR-Cas/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/metabolismo
5.
Proc Natl Acad Sci U S A ; 120(39): e2308079120, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37733743

RESUMO

TAK1 is a key modulator of both NF-κB signaling and RIPK1. In TNF signaling pathway, activation of TAK1 directly mediates the phosphorylation of IKK complex and RIPK1. In a search for small molecule activators of RIPK1-mediated necroptosis, we found R406/R788, two small molecule analogs that could promote sustained activation of TAK1. Treatment with R406 sensitized cells to TNF-mediated necroptosis and RIPK1-dependent apoptosis by promoting sustained RIPK1 activation. Using click chemistry and multiple biochemical binding assays, we showed that treatment with R406 promotes the activation of TAK1 by directly binding to TAK1, independent of its original target Syk kinase. Treatment with R406 promoted the ubiquitination of TAK1 and the interaction of activated TAK1 with ubiquitinated RIPK1. Finally, we showed that R406/R788 could promote the cancer-killing activities of TRAIL in vitro and in mouse models. Our studies demonstrate the possibility of developing small molecule TAK1 activators to potentiate the effect of TRAIL as anticancer therapies.


Assuntos
Apoptose , Neoplasias , Animais , Camundongos , Morte Celular , Citosol , Neoplasias/tratamento farmacológico , Neoplasias/genética , Ubiquitinação
6.
J Thromb Haemost ; 21(5): 1336-1351, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36792011

RESUMO

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) infection causes an emerging hemorrhagic fever in East Asia with a high mortality rate. Thrombocytopenia is a consistent feature of SFTS illness, but the mechanism remains elusive. OBJECTIVES: We aimed to better understand the role of platelets in the pathophysiology of SFTSV infection, including the development of thrombocytopenia. METHODS: Using platelets from healthy volunteers and patients with SFTS, we evaluated the functional changes in platelets against SFTSV infection. We investigated the direct effect of glycoprotein VI on platelet-SFTSV interaction by quantitative real-time PCR, molecular docking, surface plasmon resonance spectrometry, flow cytometry, western blot, and platelet functional studies in vitro. Interactions of SFTSV and platelet-SFTSV complexes with macrophages were also determined by scanning electron microscope, quantitative real-time PCR, and flow cytometry. RESULTS: This study is the first to demonstrate that platelets are capable of harboring and producing SFTSV particles. Structural and functional studies found that SFTSVs bind platelet glycoprotein VI to potentiate platelet activation, including platelet aggregation, adenosine triphosphate release, spreading, clot retraction, coagulation, phosphatidylserine exposure, thrombus formation, and adherence. In vitro mechanistic studies highlighted that the interaction of platelets with human THP-1 cells promoted SFTSV clearance and suppressed cytokine production in macrophages. However, unwanted SFTSV replication in macrophages reciprocally aggravated SFTSV persistence in the circulation, which may contribute to thrombocytopenia and other complications during SFTSV infection. CONCLUSION: These findings together highlighted the pathophysiological role of platelets in initial intrinsic defense against SFTSV infections, as well as intertwined processes with host immunity, which can also lead to thrombocytopenia and poor prognosis.


Assuntos
Infecções por Bunyaviridae , Febre Grave com Síndrome de Trombocitopenia , Trombocitopenia , Humanos , Plaquetas , Febre Grave com Síndrome de Trombocitopenia/complicações , Infecções por Bunyaviridae/complicações , Simulação de Acoplamento Molecular , Trombocitopenia/complicações , Ativação Plaquetária
7.
Stem Cell Res ; 67: 103040, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36796252

RESUMO

The transient receptor potential cation channel subfamily M member 8 (TRPM8) is a kind of non-selective cation channel which controls Ca2+ homeostasis. Mutations in TRPM8 were related to dry eye diseases (DED). Here we constructed a TRPM8 knockout cell line WAe009-A-A from the original embryonic stem cell line H9 using CRISPR/Cas9 technology, which maybe helpful for exploring the pathogenesis of DED. WAe009-A-A cells possess stem cell morphology and pluripotency as well as normal karyotype, and have the ability of differentiating into three germ layers in vitro.


Assuntos
Células-Tronco Embrionárias Humanas , Humanos , Linhagem Celular , Sistemas CRISPR-Cas , Células-Tronco Embrionárias Humanas/metabolismo , Cariótipo
8.
J Med Chem ; 66(4): 2608-2621, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36723914

RESUMO

There is growing interest in covalent targeted inhibitors in drug discovery against previously "undruggable" sites and targets. These molecules typically feature an electrophilic warhead that reacts with nucleophilic groups of protein residues, most notably the thiol group of cysteines. One main challenge in the field is to develop versatile utilizable warheads. Here, we characterize the unique features of novel arsenous warheads for reaction with thiol species in a reversible manner and further demonstrate that organoarsenic probes can be chemically tuned toward specific molecular targets by developing selective and potent inhibitors of pyruvate kinase M2 (PKM2). We show that compound 24 is a covalent and allosteric inhibitor of PKM2 and its orally bioavailable prodrug 25 exerts efficacious inhibition of PKM2-dependent tumor growth in vitro and in vivo. Our results introduce 25 and its derivatives as useful pharmacological tools and provide a general road map for targeting the protein cysteinome using arsenous warheads.


Assuntos
Descoberta de Drogas , Piruvato Quinase , Cisteína/química
9.
J Am Heart Assoc ; 11(21): e026174, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36314496

RESUMO

Background Restenosis is one of the main bottlenecks in restricting the further development of cardiovascular interventional therapy. New signaling molecules involved in the progress have continuously been discovered; however, the specific molecular mechanisms remain unclear. MTMR14 (myotubularin-related protein 14) is a novel phosphoinositide phosphatase that has a variety of biological functions and is involved in diverse biological processes. However, the role of MTMR14 in vascular biology remains unclear. Herein, we addressed the role of MTMR14 in neointima formation and vascular smooth muscle cell (VSMC) proliferation after vessel injury. Methods and Results Vessel injury models were established using SMC-specific conditional MTMR14-knockout and -transgenic mice. Neointima formation was assessed by histopathological methods, and VSMC proliferation and migration were assessed using fluorescence ubiquitination-based cell cycle indicator, transwell, and scratch wound assay. Neointima formation and the expression of MTMR14 was increased after injury. MTMR14 deficiency accelerated neointima formation and promoted VSMC proliferation after injury, whereas MTMR14 overexpression remarkably attenuated this process. Mechanistically, we demonstrated that MTMR14 suppressed the activation of PLK1 (polo-like kinase 1) by interacting with it, which further leads to the inhibition of the activation of MEK/ERK/AKT (mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase/protein kinase B), thereby inhibiting the proliferation of VSMC from the medial to the intima and thus preventing neointima formation. Conclusions MTMR14 prevents neointima formation and VSMC proliferation by inhibiting PLK1. Our findings reveal that MTMR14 serves as an inhibitor of VSMC proliferation and establish a link between MTMR14 and PLK1 in regulating VSMC proliferation. MTMR14 may become a novel potential therapeutic target in the treatment of restenosis.


Assuntos
Monoéster Fosfórico Hidrolases , Proteínas Serina-Treonina Quinases , Lesões do Sistema Vascular , Animais , Camundongos , Movimento Celular , Proliferação de Células , Células Cultivadas , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Neointima/patologia , Monoéster Fosfórico Hidrolases/metabolismo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/prevenção & controle , Lesões do Sistema Vascular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Quinase 1 Polo-Like
11.
Nat Commun ; 13(1): 2529, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35534502

RESUMO

Hypoxia-inducible factors (HIFs) are α/ß heterodimeric transcription factors modulating cellular responses to the low oxygen condition. Among three HIF-α isoforms, HIF-3α is the least studied to date. Here we show that oleoylethanolamide (OEA), a physiological lipid known to regulate food intake and metabolism, binds selectively to HIF-3α. Through crystallographic analysis of HIF-3 α/ß heterodimer in both apo and OEA-bound forms, hydrogen-deuterium exchange mass spectrometry (HDX-MS), molecular dynamics (MD) simulations, and biochemical and cell-based assays, we unveil the molecular mechanism of OEA entry and binding to the PAS-B pocket of HIF-3α, and show that it leads to enhanced heterodimer stability and functional modulation of HIF-3. The identification of HIF-3α as a selective lipid sensor is consistent with recent human genetic findings linking HIF-3α with obesity, and demonstrates that endogenous metabolites can directly interact with HIF-α proteins to modulate their activities, potentially as a regulatory mechanism supplementary to the well-known oxygen-dependent HIF-α hydroxylation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas Repressoras , Proteínas Reguladoras de Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Endocanabinoides , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Ligantes , Ácidos Oleicos , Oxigênio/metabolismo
12.
Angew Chem Int Ed Engl ; 61(17): e202115472, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35199422

RESUMO

The control of endogenous protein activity with light inside live cells is helpful for the high spatiotemporal probing of their dynamic roles. Herein, we report the first small-molecule-ligand-directed caging approach to control the endogenous human O6 -alkylguanine-DNA alkyltransferase (AGT) activity with light, and the caged AGT is constructed from the native intracellular AGT. The photo-responsive O6 -benzylguanine derivative O6 -NBG3 is developed to site-specifically cage the AGT's catalytic cysteine residue, and the light irradiation on-demand restores AGT's activity in vitro, in bacteria, and in mammalian cells. With O6 -NBG3, the alkylated AGT is dealkylated for the first time to recover the DNA repair activity in breast cancer MCF-7 cells by the dose-dependent light irradiation. This decaging strategy enables the localized modulation of endogenous AGT activity with high temporal precision without genetic engineering, which holds great potential for therapeutic applications.


Assuntos
Alquil e Aril Transferases , Guanina , Animais , Guanina/química , Humanos , Ligantes , Mamíferos/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/química
13.
Stem Cell Res ; 58: 102622, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929444

RESUMO

A 25-years-old hypertrophic cardiomyopathy male patient donated his peripheral blood mononuclear cells (PBMCs) with heterozygote mutation in theTNNT2 gene. We generated induced pluripotent stem cell (iPSC) with normal karyotypic and expressing NANOG, Lin28, GDF3 and DNMT3. The iPSC line has demonstrated pluripotency by differentiating into three germ layers in vitro. The ZZUNEUi021-A would serve as an in vitro model for loss of TNNT2 function.


Assuntos
Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Adulto , Cardiomiopatia Hipertrófica/genética , Diferenciação Celular , Genes Homeobox , Humanos , Leucócitos Mononucleares , Masculino , Mutação/genética , Troponina T/genética
14.
Signal Transduct Target Ther ; 6(1): 378, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34732694

RESUMO

The current COVID-19 pandemic has heavily burdened the global public health system and may keep simmering for years. The frequent emergence of immune escape variants have spurred the search for prophylactic vaccines and therapeutic antibodies that confer broad protection against SARS-CoV-2 variants. Here we show that the bivalency of an affinity maturated fully human single-domain antibody (n3113.1-Fc) exhibits exquisite neutralizing potency against SARS-CoV-2 pseudovirus, and confers effective prophylactic and therapeutic protection against authentic SARS-CoV-2 in the host cell receptor angiotensin-converting enzyme 2 (ACE2) humanized mice. The crystal structure of n3113 in complex with the receptor-binding domain (RBD) of SARS-CoV-2, combined with the cryo-EM structures of n3113 and spike ecto-domain, reveals that n3113 binds to the side surface of up-state RBD with no competition with ACE2. The binding of n3113 to this novel epitope stabilizes spike in up-state conformations but inhibits SARS-CoV-2 S mediated membrane fusion, expanding our recognition of neutralization by antibodies against SARS-CoV-2. Binding assay and pseudovirus neutralization assay show no evasion of recently prevalent SARS-CoV-2 lineages, including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), and Delta (B.1.617.2) for n3113.1-Fc with Y58L mutation, demonstrating the potential of n3113.1-Fc (Y58L) as a promising candidate for clinical development to treat COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/química , Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , COVID-19 , SARS-CoV-2/química , Anticorpos de Cadeia Única/química , Enzima de Conversão de Angiotensina 2/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Cristalografia por Raios X , Epitopos/química , Epitopos/imunologia , Humanos , Camundongos , SARS-CoV-2/imunologia , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/uso terapêutico
15.
Exp Hematol ; 103: 52-59.e2, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34474146

RESUMO

Since the use of Bruton's tyrosine kinase (BTK) inhibitor ibrutinib in relapsed/refractory (R/R) mantle cell lymphoma (MCL), the problem of drug resistance has become increasingly prominent. Though it has been proven that the nonclassic nuclear factor κB pathway (nonclassic NF-κB pathway) correlates with ibrutinib resistance in MCL, the upstream regulator is unknown. In the present study, conserved helix-loop-helix ubiquitous kinase (CHUK) overexpression accelerated proliferation and suppressed apoptosis of MCL cells after ibrutinib treatment in vitro. The results of luciferase reporter assay, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blot revealed that CHUK was targeted and negatively regulated by miRNA-223-3p. miRNA-223-3p knockdown promoted proliferation and inhibited apoptosis of MCL cells after ibrutinib treatment in vitro and vivo, whereas CHUK knockdown reversed downregulated miRNA-223-3p-promoted cell proliferation after ibrutinib treatment in vitro. In conclusion, miRNA-223-3p modulates ibrutinib resistance through regulation of the CHUK/NF-κB signaling pathway in MCL, which is crucial in providing a marker to predict disease response.


Assuntos
Adenina/análogos & derivados , Linfoma de Célula do Manto/tratamento farmacológico , MicroRNAs/genética , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Adenina/farmacologia , Adenina/uso terapêutico , Animais , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/metabolismo , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia
16.
Nutr Metab (Lond) ; 18(1): 71, 2021 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-34256802

RESUMO

BACKGROUND: Cardiac hypertrophy is an independent risk factor of many cardiovascular diseases. Studies have demonstrated that microRNA-126 (miR-126) was involved in angiogenesis during physiological and pathological process. However, its role in cardiac hypertrophy has not been known clearly. Our previous study demonstrated that asiatic acid (AA) has obvious protective effect on cardiac hypertrophy. Here, this study aimed to discover the regulatory role of miR-126 and its mechanism in cardiac hypertrophy, and to determine whether AA's anti-hypertrophy effect is partially miR-126 dependent. METHODS: Male Sprague Dawley rats were AngII infused via osmotic minipumps for 4 weeks and were treated with AA (20 mg/kg/day) by oral gavage. Cardiac hypertrophy was assessed using the echocardiography and histological analysis. In vitro studies,cardiomyocyte and cardiac fibroblasts (CF) were treted with AngII and AngII plus AA. And, the effect of AA on miR-126 and PI3K/AKT signaling pathway was investigated. RESULTS: Treatment of rats with AA decreased the ratio of heart weight to tibia length and hypertrophy markers. In vitro exprements demonstrated that AA significantly attenuated AngII-induced cardiac growth and cardiac fibroblast collagen expression. Moreover, our results found downregulation of miR-126 and activation of PI3K/AKT signaling pathway in AngII infusion induced cardiac hypertrophy model. It was also determined that miR-126 targets PIK3R2 directly. CONCLUSIONS: AA supplementation upregulated the expression of miR-126 and conferred cardio-protection effect against AngII induced cardiac hypertrophy.

17.
Nat Commun ; 12(1): 4635, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330908

RESUMO

SARS-CoV-2, the causative agent of COVID-191, features a receptor-binding domain (RBD) for binding to the host cell ACE2 protein1-6. Neutralizing antibodies that block RBD-ACE2 interaction are candidates for the development of targeted therapeutics7-17. Llama-derived single-domain antibodies (nanobodies, ~15 kDa) offer advantages in bioavailability, amenability, and production and storage owing to their small sizes and high stability. Here, we report the rapid selection of 99 synthetic nanobodies (sybodies) against RBD by in vitro selection using three libraries. The best sybody, MR3 binds to RBD with high affinity (KD = 1.0 nM) and displays high neutralization activity against SARS-CoV-2 pseudoviruses (IC50 = 0.42 µg mL-1). Structural, biochemical, and biological characterization suggests a common neutralizing mechanism, in which the RBD-ACE2 interaction is competitively inhibited by sybodies. Various forms of sybodies with improved potency have been generated by structure-based design, biparatopic construction, and divalent engineering. Two divalent forms of MR3 protect hamsters from clinical signs after live virus challenge and a single dose of the Fc-fusion construct of MR3 reduces viral RNA load by 6 Log10. Our results pave the way for the development of therapeutic nanobodies against COVID-19 and present a strategy for rapid development of targeted medical interventions during an outbreak.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , SARS-CoV-2/imunologia , Anticorpos de Domínio Único/imunologia , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/ultraestrutura , Anticorpos Antivirais/farmacologia , Anticorpos Antivirais/ultraestrutura , Sítios de Ligação/imunologia , COVID-19/prevenção & controle , COVID-19/virologia , Microscopia Crioeletrônica , Cristalografia por Raios X , Feminino , Humanos , Espectrometria de Massas/métodos , Mesocricetus , Camundongos Endogâmicos C57BL , Testes de Neutralização , Ligação Proteica/efeitos dos fármacos , Receptores Virais/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo
18.
J Cell Mol Med ; 25(16): 7867-7877, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34190420

RESUMO

Cardiac hypertrophy and the resultant heart failure are among the most common causes of morbidity and mortality worldwide; thus, identifying the key factor mediating pathological cardiac hypertrophy is critically important for developing the strategy to protect against heart failure. Runx1 (Runt-related transcription factor 1) acts as an essential transcription factor that functions in a variety of cellular processes including differentiation, proliferation, tissue growth and DNA damage response. However, relatively little is known about the role of Runx1 in heart, especially cardiac hypertrophy and heart failure. In the present study, we investigated the role of Runx1 in experimentally pathological cardiac hypertrophy. The in vitro model was induced by Ang II exposure to cultured neonatal rat cardiomyocytes, and the in vivo pathological cardiac hypertrophy models were induced by chronic pressure overload in mice. Runx1 expression is increased in heart tissues from mice with pressure overload-induced cardiac hypertrophy and in neonatal rat cardiomyocytes in response to Ang II stimulation. Moreover, knockdown of cardiac Runx1 alleviates the pressure overload-induced cardiac hypertrophy. Mechanistically, Runx1 activates the p53 signalling by binding to the p53 gene and promotes its transcription. Rescue experiments indicate that Runx1 promotes cardiac hypertrophy in a p53-dependent manner. Remarkably, we demonstrated that Ro5-3335 (a Runx1 inhibitor) acts as a potential therapeutic drug for treating pathological cardiac hypertrophy. In summary, we conclude that Runx1 is a novel mediator and therapeutic target for pathological cardiac hypertrophy.


Assuntos
Cardiomegalia/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Miócitos Cardíacos/patologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Miócitos Cardíacos/metabolismo , Ratos , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
19.
Cell Discov ; 7(1): 41, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34075030

RESUMO

RIPK1, a death domain-containing kinase, has been recognized as an important therapeutic target for inhibiting apoptosis, necroptosis, and inflammation under pathological conditions. RIPK1 kinase inhibitors have been advanced into clinical studies for the treatment of various human diseases. One of the current bottlenecks in developing RIPK1 inhibitors is to discover new approaches to inhibit this kinase as only limited chemotypes have been developed. Here we describe Necrostatin-34 (Nec-34), a small molecule that inhibits RIPK1 kinase with a mechanism distinct from known RIPK1 inhibitors such as Nec-1s. Mechanistic studies suggest that Nec-34 stabilizes RIPK1 kinase in an inactive conformation by occupying a distinct binding pocket in the kinase domain. Furthermore, we show that Nec-34 series of compounds can synergize with Nec-1s to inhibit RIPK1 in vitro and in vivo. Thus, Nec-34 defines a new strategy to target RIPK1 kinase and provides a potential option of combinatorial therapy for RIPK1-mediated diseases.

20.
Nucleic Acids Res ; 49(7): 3796-3813, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33744966

RESUMO

The family of Poly(A)-binding proteins (PABPs) regulates the stability and translation of messenger RNAs (mRNAs). Here we reported that the three members of PABPs, including PABPC1, PABPC3 and PABPC4, were identified as novel substrates for MKRN3, whose deletion or loss-of-function mutations were genetically associated with human central precocious puberty (CPP). MKRN3-mediated ubiquitination was found to attenuate the binding of PABPs to the poly(A) tails of mRNA, which led to shortened poly(A) tail-length of GNRH1 mRNA and compromised the formation of translation initiation complex (TIC). Recently, we have shown that MKRN3 epigenetically regulates the transcription of GNRH1 through conjugating poly-Ub chains onto methyl-DNA bind protein 3 (MBD3). Therefore, MKRN3-mediated ubiquitin signalling could control both transcriptional and post-transcriptional switches of mammalian puberty initiation. While identifying MKRN3 as a novel tissue-specific translational regulator, our work also provided new mechanistic insights into the etiology of MKRN3 dysfunction-associated human CPP.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Proteínas de Ligação a Poli(A)/metabolismo , Precursores de Proteínas/genética , Puberdade Precoce , RNA Mensageiro/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Puberdade Precoce/genética , Puberdade Precoce/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...