Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
2.
Gut Microbes ; 16(1): 2334970, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38563680

RESUMO

Gastrointestinal (GI) infection is evidenced with involvement in COVID-19 pathogenesis caused by SARS-CoV-2. However, the correlation between GI microbiota and the distinct pathogenicity of SARS-CoV-2 Proto and its emerging variants remains unclear. In this study, we aimed to determine if GI microbiota impacted COVID-19 pathogenesis and if the effect varied between SARS-CoV-2 Proto and its variants. We performed an integrative analysis of histopathology, microbiomics, and transcriptomics on the GI tract fragments from rhesus monkeys infected with SARS-CoV-2 proto or its variants. Based on the degree of pathological damage and microbiota profile in the GI tract, five of SARS-CoV-2 strains were classified into two distinct clusters, namely, the clusters of Alpha, Beta and Delta (ABD), and Proto and Omicron (PO). Notably, the abundance of potentially pathogenic microorganisms increased in ABD but not in the PO-infected rhesus monkeys. Specifically, the high abundance of UCG-002, UCG-005, and Treponema in ABD virus-infected animals positively correlated with interleukin, integrins, and antiviral genes. Overall, this study revealed that infection-induced alteration of GI microbiota and metabolites could increase the systemic burdens of inflammation or pathological injury in infected animals, especially in those infected with ABD viruses. Distinct GI microbiota and metabolite profiles may be responsible for the differential pathological phenotypes of PO and ABD virus-infected animals. These findings improve our understanding the roles of the GI microbiota in SARS-CoV-2 infection and provide important information for the precise prevention, control, and treatment of COVID-19.


Assuntos
COVID-19 , Microbioma Gastrointestinal , Animais , SARS-CoV-2 , Virulência , Macaca mulatta
3.
Cell Signal ; 120: 111191, 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38685521

RESUMO

Recent studies have revealed that PTPRZ1-MET (ZM) fusion plays a pivotal role in the progression of glioma to glioblastoma multiforme (GBM), thus serving as a biomarker to distinguish between primary GBM and secondary GBM (sGBM). However, the mechanisms through which ZM fusion influences this progression remain to be elucidated. GBMs with ZM showed poorer prognoses and greater infiltration of tumor-associated macrophages (TAMs) than those without ZM. Glioma stem-like cells (GSCs) and TAMs play complex roles in glioma recurrence, glioma progression and therapy resistance. In this study, we analyzed RNA-seq data from sGBM patients' glioma tissues with or without ZM fusion, and found that stemness and macrophage markers were more highly expressed in sGBM patients harboring ZM than in those without ZM fusion. ZM enhanced the self-renewal and proliferation of GSCs, thereby accelerating glioma progression. In addition, ZM-positive GSCs facilitated the infiltration of TAMs and drove their polarization toward an immunosuppressive phenotype, which was primarily accomplished through the extracellular secretion of ISG20. Our research identified the MET-STAT3-ISG20 axis within GSCs, thus demonstrating the critical role of ZM in GBM initiation and progression. Our study demonstrated that, in contrast to ZM-positive differentiated glioma cells, ZM-positive GSCs upregulated ISG20 expression through the MET-STAT3-ISG20 axis. The extracellular secretion of ISG20 recruited and induced M2-like polarization in macrophages, thereby promoting tumor progression. Our results reveal a novel mechanism involved in ZM-positive GBM pathogenesis and identify potential therapeutic targets.

4.
Biochem Biophys Res Commun ; 704: 149706, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38432144

RESUMO

Glioma patients often undertake psychiatric disorders such as depression and anxiety. There are several clinical epidemiological studies on glioma-associated depression, but basic research and corresponding animal experiments are still lacking. Here, we observed that glioma-bearing mice exhibited atypical depression-like behaviors in orthotopic glioma mouse models. The concentrations of monoamine neurotransmitters were detected by enzyme-linked immunosorbent assay (ELISA), revealing a decrease in 5-hydroxytryptamine (5-HT) levels in para-glioma tissues. The related gene expression levels also altered, detected by quantitative RT-PCR. Then, we developed a glioma-depression comorbidity mouse model. Through sucrose preference test (SPT), forced swimming test (FST), tail suspension test (TST) and other tests, we found that the occurrence of glioma could lead to changes in depression-like behaviors in a chronic unpredictable mild stress (CUMS) mouse model. The results of RNA sequencing (RNA-seq) indicated that the altered expression of glutamatergic synapse related genes in the paratumor tissues might be one of the main molecular features of the comorbidity model. Our findings suggested that the presence of glioma caused and altered depression-like behaviors, which was potentially related to the 5-HT and glutamatergic synapse pathways.


Assuntos
Depressão , Serotonina , Humanos , Camundongos , Animais , Depressão/metabolismo , Serotonina/metabolismo , Antidepressivos/farmacologia , Comportamento Animal , Natação , Estresse Psicológico/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo
5.
Elife ; 122024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38415809

RESUMO

Despite intense research on mice, the transcriptional regulation of neocortical neurogenesis remains limited in humans and non-human primates. Cortical development in rhesus macaque is known to recapitulate multiple facets of cortical development in humans, including the complex composition of neural stem cells and the thicker supragranular layer. To characterize temporal shifts in transcriptomic programming responsible for differentiation from stem cells to neurons, we sampled parietal lobes of rhesus macaque at E40, E50, E70, E80, and E90, spanning the full period of prenatal neurogenesis. Single-cell RNA sequencing produced a transcriptomic atlas of developing parietal lobe in rhesus macaque neocortex. Identification of distinct cell types and neural stem cells emerging in different developmental stages revealed a terminally bifurcating trajectory from stem cells to neurons. Notably, deep-layer neurons appear in the early stages of neurogenesis, while upper-layer neurons appear later. While these different lineages show overlap in their differentiation program, cell fates are determined post-mitotically. Trajectories analysis from ventricular radial glia (vRGs) to outer radial glia (oRGs) revealed dynamic gene expression profiles and identified differential activation of BMP, FGF, and WNT signaling pathways between vRGs and oRGs. These results provide a comprehensive overview of the temporal patterns of gene expression leading to different fates of radial glial progenitors during neocortex layer formation.


Assuntos
Neocórtex , Células-Tronco Neurais , Feminino , Gravidez , Animais , Camundongos , Transcriptoma , Macaca mulatta , Perfilação da Expressão Gênica
6.
Proc Natl Acad Sci U S A ; 121(10): e2317026121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38408250

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been detected in almost all organs of coronavirus disease-19 patients, although some organs do not express angiotensin-converting enzyme-2 (ACE2), a known receptor of SARS-CoV-2, implying the presence of alternative receptors and/or co-receptors. Here, we show that the ubiquitously distributed human transferrin receptor (TfR), which binds to diferric transferrin to traffic between membrane and endosome for the iron delivery cycle, can ACE2-independently mediate SARS-CoV-2 infection. Human, not mouse TfR, interacts with Spike protein with a high affinity (KD ~2.95 nM) to mediate SARS-CoV-2 endocytosis. TfR knock-down (TfR-deficiency is lethal) and overexpression inhibit and promote SARS-CoV-2 infection, respectively. Humanized TfR expression enables SARS-CoV-2 infection in baby hamster kidney cells and C57 mice, which are known to be insusceptible to the virus infection. Soluble TfR, Tf, designed peptides blocking TfR-Spike interaction and anti-TfR antibody show significant anti-COVID-19 effects in cell and monkey models. Collectively, this report indicates that TfR is a receptor/co-receptor of SARS-CoV-2 mediating SARS-CoV-2 entry and infectivity by likely using the TfR trafficking pathway.


Assuntos
COVID-19 , Animais , Humanos , Camundongos , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Peptidil Dipeptidase A/metabolismo , Ligação Proteica , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo
7.
Signal Transduct Target Ther ; 9(1): 32, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38351062

RESUMO

The appropriate and specific response of nerve cells to various external cues is essential for the establishment and maintenance of neural circuits, and this process requires the proper recruitment of adaptor molecules to selectively activate downstream pathways. Here, we identified that DOK6, a member of the Dok (downstream of tyrosine kinases) family, is required for the maintenance of peripheral axons, and that loss of Dok6 can cause typical peripheral neuropathy symptoms in mice, manifested as impaired sensory, abnormal posture, paw deformities, blocked nerve conduction, and dysmyelination. Furthermore, Dok6 is highly expressed in peripheral neurons but not in Schwann cells, and genetic deletion of Dok6 in peripheral neurons led to typical peripheral myelin outfolding, axon destruction, and hindered retrograde axonal transport. Specifically, DOK6 acts as an adaptor protein for selectivity-mediated neurotrophic signal transduction and retrograde transport for TrkC and Ret but not for TrkA and TrkB. DOK6 interacts with certain proteins in the trafficking machinery and controls their phosphorylation, including MAP1B, Tau and Dynein for axonal transport, and specifically activates the downstream ERK1/2 kinase pathway to maintain axonal survival and homeostasis. This finding provides new clues to potential insights into the pathogenesis and treatment of hereditary peripheral neuropathies and other degenerative diseases.


Assuntos
Doenças do Sistema Nervoso Periférico , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Axônios/metabolismo , Axônios/patologia , Neurônios/metabolismo , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Transdução de Sinais/genética
8.
J Control Release ; 366: 479-493, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38184234

RESUMO

mRNA-based vaccines and therapeutic agents hold great promise in prevention and treatment of human diseases, yet high percentage of systemic adverse effect in clinic remains a big safety concern. One major potential cause is a high level of leakage of the locally inoculated mRNA vaccine nanoparticles into circulation. We have screened and optimized a core-shell structured lipopolyplex (LPP) formulation for mRNA with a tissue-retention property. Upon intramuscular inoculation, the mRNA-encapsulated LPP nanoparticles were preferentially taken up by the phagocytic antigen-presentation cells, and potently promoted dendritic cell maturation. We applied the new formulation to prepare a prophylactic vaccine for SARS-CoV-2, and observed potent humoral and cellular immune responses from the vaccine in both murine models and non-human primates. More importantly, the vaccine demonstrated a benign safety profile in non-human primates, with limited side effects after repeated treatment with high dosages of LPP/mRNA. Taken together, the inoculation site-retained vaccine formulation serves as a promising vehicle for mRNA vaccines and therapeutic agents.


Assuntos
COVID-19 , Vacinas de mRNA , Humanos , Animais , Camundongos , SARS-CoV-2/genética , Vacinas contra COVID-19 , COVID-19/prevenção & controle , Apresentação de Antígeno , RNA Mensageiro , Primatas , Anticorpos Antivirais , Anticorpos Neutralizantes
9.
iScience ; 27(2): 108706, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38288355

RESUMO

Sirt6, a member of the class III histone deacetylases (HDACs), functions in the regulation of genomic stability, DNA repair, cancer, metabolism and aging. Sirt6 deficiency is lethal, and newborn SIRT6-null cynomolgus monkeys show unfinished brain development. After the generation of a cortex-specific Sirt6 conditional knockout mouse model, we investigated the specific deletion of Sirt6 in NPCs at E10.5. This study found that Sirt6 deficiency causes excessive proliferation of neural precursor cells (NPCs) and retards differentiation. The results suggest that endogenous Sirt6 in NPCs regulates histone acetylation and limits stemness-related genes, including Notch1, in order to participate in NPC fate determination. These findings help elucidate Sirt6's role in brain development and in NPC fate determination while providing data on species generality and differentiation.

10.
Trends Mol Med ; 30(2): 110-112, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38092623

RESUMO

Diffuse intrinsic pontine glioma (DIPG) remains intractable to conventional treatments. While targeted therapy and immuno-oncology advances offer hope, few strategies show promising results. In a recent article in Cancer Cell, Ausejo-Mauleon et al. introduce TIM-3 blockade as a potential breakthrough for DIPG treatment by targeting cancer cells and regulating the immune microenvironment simultaneously.


Assuntos
Neoplasias do Tronco Encefálico , Glioma , Humanos , Glioma/tratamento farmacológico , Neoplasias do Tronco Encefálico/terapia , Receptor Celular 2 do Vírus da Hepatite A , Microambiente Tumoral
11.
Orthop Surg ; 16(1): 111-122, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38044447

RESUMO

OBJECTIVE: With advancements in minimally invasive techniques, the use of spinal fusion surgery is rapidly increasing and transfusion rates are decreasing. Routine preoperative ABO/Rh blood type and antibody screening (T&S) laboratory tests may not be appropriate for all spinal fusion patients. Herein, we constructed a nomogram to assess patient transfusion risk based on various risk factors in patients undergoing spinal fusion surgery, so that preoperative T&S testing can be selectively scheduled in appropriate patients to reduce healthcare and patient costs. METHODS: Patients who underwent spinal fusion surgery between 01/2020 and 03/2023 were retrospectively examined and classified into the training (n = 3533, 70%) and validation (n = 1515, 30%) datasets. LASSO and multivariable logistic regression were used to analyze risk factors for blood transfusion. Nomogram predictive model was built according to the independent predictors and mode predictive power was validated using consistency index (C-index), Hosmer-Lemeshow (HL) test, calibration curve analysis and area under the curve (AUC) for receiver operating characteristic (ROC) curve. Bootstrap resampling was used for internal validation. Decision curve analysis (DCA) was applied to evaluate the model's performance in the clinic. RESULTS: Being female, age, BMI, admission route, critical patient, operative time, heart failure, end-stage renal disease or chronic kidney disease (ESRD or CKD), anemia, and coagulation defect were predictors of blood transfusion for spinal fusion. A prediction nomogram was developed according to a multivariate model with good discriminatory power (C-index = 0.887); Bootstrap resampling internal validation C-index was 0.883. Calibration curves showed strong matching between the predicted and actual probabilities of the training and validation sets. HL tests for the training and validation sets had p-values of 0.327 and 0.179, respectively, indicating good calibration. When applied to the training set, the following parameters were found: AUC: 0.895, 95% CI: 0.871-0.919, sensitivity 78.2%, specificity 86.7%, positive predictive value 29.4% and negative predictive value 98.2%. If the model were applied in the training set, 2911 T&S tests (82.4%) would be eliminated, equaling a RMB349,320 cost reduction. The AUC in the internal validation was: 0.879, 95% CI: 0.839-0.927, sensitivity 75.2%, specificity 88.8%, positive predictive value 34.3%, negative predictive value 97.9%, would eliminate 1276 T&S tests (84.2%), saving RMB 153,120. The DCA curve indicated good clinical application value. CONCLUSION: The nomogram based on 10 independent factors can help healthcare professionals predict the risk of transfusion for patients undergoing spinal fusion surgery to target preoperative T&S testing to appropriate patients and reduce healthcare costs.


Assuntos
Nomogramas , Fusão Vertebral , Humanos , Feminino , Masculino , Estudos Retrospectivos , Hospitalização
12.
FEBS Open Bio ; 14(1): 138-147, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37953466

RESUMO

Extracellular vesicles (EV), important messengers in intercellular communication, can load and transport various bioactive components and participate in different biological processes. We previously isolated glioma human endothelial cells (GhECs) and found that GhECs, rather than normal human brain endothelial cells (NhECs), exhibit specific enrichment of MYO1C into EVs and promote the migration of glioma cells. In this study, we explored the mechanism by which MYO1C is secreted into EVs. We report that such secretion is dependent on RAB31, RAB27B, and FAS. When expression of RAB31 increases, MYO1C is enriched in secretory EVs. Finally, we identified an EV export mechanism for MYO1C that promotes glioma cell invasion and is dependent on RAB31 in GhECs. In summary, our data indicate that the knockdown of RAB31 can reduce enrichment of MYO1C in extracellular vesicles, thereby attenuating the promotion of glioma cell invasion by GhEC-EVs.


Assuntos
Vesículas Extracelulares , Glioma , Humanos , Células Endoteliais/metabolismo , Glioma/genética , Glioma/metabolismo , Transporte Biológico , Vesículas Extracelulares/metabolismo , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
13.
MedComm (2020) ; 4(6): e432, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38020713

RESUMO

Immune responses induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection play a critical role in the pathogenesis and outcome of coronavirus disease 2019 (COVID-19). However, the dynamic profile of immune responses postinfection by SARS-CoV-2 variants of concern (VOC) is not fully understood. In this study, peripheral blood mononuclear cells single-cell sequencing was performed to determine dynamic profiles of immune response to Prototype, Alpha, Beta, and Delta in a rhesus monkey model. Overall, all strains induced dramatic changes in both cellular subpopulations and gene expression levels at 1 day postinfection (dpi), which associated function including adaptive immune response, innate immunity, and IFN response. COVID-19-related genes revealed different gene profiles at 1 dpi among the four SARS-CoV-2 strains, including genes reported in COVID-19 patients with increased risk of autoimmune disease and rheumatic diseases. Delta-infected animal showed inhibition of translation pathway. B cells, T cells, and monocytes showed much commonality rather than specificity among the four strains. Monocytes were the major responders to SARS-CoV-2 infection, and the response lasted longer in Alpha than the other strains. Thus, this study reveals the early immune responses induced by SARS-CoV-2 Proto or its variants in nonhuman primates, which is important information for controlling rapidly evolving viruses.

14.
Signal Transduct Target Ther ; 8(1): 424, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37935665

RESUMO

Glioma is the most prevalent brain tumor, presenting with limited treatment options, while patients with malignant glioma and glioblastoma (GBM) have poor prognoses. The physical obstacle to drug delivery imposed by the blood‒brain barrier (BBB) and glioma stem cells (GSCs), which are widely recognized as crucial elements contributing to the unsatisfactory clinical outcomes. In this study, we found a small molecule, gambogic amide (GA-amide), exhibited the ability to effectively penetrate the blood-brain barrier (BBB) and displayed a notable enrichment within the tumor region. Moreover, GA-amide exhibited significant efficacy in inhibiting tumor growth across various in vivo glioma models, encompassing transgenic and primary patient-derived xenograft (PDX) models. We further performed a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) knockout screen to determine the druggable target of GA-amide. By the combination of the cellular thermal shift assay (CETSA), the drug affinity responsive target stability (DARTS) approach, molecular docking simulation and surface plasmon resonance (SPR) analysis, WD repeat domain 1 (WDR1) was identified as the direct binding target of GA-amide. Through direct interaction with WDR1, GA-amide promoted the formation of a complex involving WDR1, MYH9 and Cofilin, which accelerate the depolymerization of F-actin to inhibit the invasion of patient-derived glioma cells (PDCs) and induce PDC apoptosis via the mitochondrial apoptotic pathway. In conclusion, our study not only identified GA-amide as an effective and safe agent for treating glioma but also shed light on the underlying mechanisms of GA-amide from the perspective of cytoskeletal homeostasis.


Assuntos
Glioma , Humanos , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Citoesqueleto , Amidas , Proteínas dos Microfilamentos/uso terapêutico
15.
EMBO J ; 42(22): e113383, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37807845

RESUMO

Notch signaling pathway activity, particularly fluctuations in the biologically active effector fragment NICD, is required for rapid and efficient dynamic regulation of proper fate decisions in stem cells. In this study, we identified NEDD4-binding protein 1 (N4BP1), which is highly expressed in the developing mouse cerebral cortex, as a negative modulator of Notch signaling dynamics in neural progenitor cells. Intriguingly, N4BP1 regulated NICD stability specifically after Notch1 S3 cleavage through ubiquitin-mediated degradation that depended on its RAM domain, not its PEST domain, as had been extensively and previously described. The CoCUN domain in N4BP1, particularly the "Phe-Pro" motif (862/863 amino acid), was indispensable for mediating NICD degradation. The Ring family E3 ligase Trim21 was, in contrast to other NEDD4 family members, required for N4BP1-regulated NICD degradation. Overexpression of N4BP1 in cortical neural progenitors promoted neural stem cell differentiation, whereas neural progenitor cells lacking N4BP1 were sensitized to Notch signaling, resulting in the maintenance of stem-like properties in neural progenitor cells and lower production of cortical neurons.


Assuntos
Neocórtex , Células-Tronco Neurais , Animais , Camundongos , Diferenciação Celular/fisiologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Receptor Notch1/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia
16.
Nat Commun ; 14(1): 5191, 2023 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-37626079

RESUMO

Many of the currently available COVID-19 vaccines and therapeutics are not effective against newly emerged SARS-CoV-2 variants. Here, we developed the metallo-enzyme domain of angiotensin converting enzyme 2 (ACE2)-the cellular receptor of SARS-CoV-2-into an IgM-like inhalable molecule (HH-120). HH-120 binds to the SARS-CoV-2 Spike (S) protein with high avidity and confers potent and broad-spectrum neutralization activity against all known SARS-CoV-2 variants of concern. HH-120 was developed as an inhaled formulation that achieves appropriate aerodynamic properties for rodent and monkey respiratory system delivery, and we found that early administration of HH-120 by aerosol inhalation significantly reduced viral loads and lung pathology scores in male golden Syrian hamsters infected by the SARS-CoV-2 ancestral strain (GDPCC-nCoV27) and the Delta variant. Our study presents a meaningful advancement in the inhalation delivery of large biologics like HH-120 (molecular weight (MW) ~ 1000 kDa) and demonstrates that HH-120 can serve as an efficacious, safe, and convenient agent against SARS-CoV-2 variants. Finally, given the known role of ACE2 in viral reception, it is conceivable that HH-120 has the potential to be efficacious against additional emergent coronaviruses.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Masculino , Animais , Cricetinae , Humanos , Vacinas contra COVID-19 , SARS-CoV-2/genética , Mesocricetus , Imunoglobulina M
17.
EMBO Rep ; 24(9): e56512, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37437058

RESUMO

Long interspersed element 1 (LINE-1) is the only active autonomous mobile element in the human genome. Its transposition can exert deleterious effects on the structure and function of the host genome and cause sporadic genetic diseases. Tight control of LINE-1 mobilization by the host is crucial for genetic stability. In this study, we report that MOV10 recruits the main decapping enzyme DCP2 to LINE-1 RNA and forms a complex of MOV10, DCP2, and LINE-1 RNP, exhibiting liquid-liquid phase separation (LLPS) properties. DCP2 cooperates with MOV10 to decap LINE-1 RNA, which causes degradation of LINE-1 RNA and thus reduces LINE-1 retrotransposition. We here identify DCP2 as one of the key effector proteins determining LINE-1 replication, and elucidate an LLPS mechanism that facilitates the anti-LINE-1 action of MOV10 and DCP2.


Assuntos
Grânulos Citoplasmáticos , RNA Helicases , Humanos , Grânulos Citoplasmáticos/metabolismo , Endorribonucleases/genética , Elementos Nucleotídeos Longos e Dispersos , RNA/metabolismo , RNA Helicases/metabolismo
18.
Genomics Proteomics Bioinformatics ; 21(5): 1014-1029, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37451436

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the persistent coronavirus disease 2019 (COVID-19) pandemic, which has resulted in millions of deaths worldwide and brought an enormous public health and global economic burden. The recurring global wave of infections has been exacerbated by growing variants of SARS-CoV-2. In this study, the virological characteristics of the original SARS-CoV-2 strain and its variants of concern (VOCs; including Alpha, Beta, and Delta) in vitro, as well as differential transcriptomic landscapes in multiple organs (lung, right ventricle, blood, cerebral cortex, and cerebellum) from the infected rhesus macaques, were elucidated. The original strain of SARS-CoV-2 caused a stronger innate immune response in host cells, and its VOCs markedly increased the levels of subgenomic RNAs, such as N, Orf9b, Orf6, and Orf7ab, which are known as the innate immune antagonists and the inhibitors of antiviral factors. Intriguingly, the original SARS-CoV-2 strain and Alpha variant induced larger alteration of RNA abundance in tissues of rhesus monkeys than Beta and Delta variants did. Moreover, a hyperinflammatory state and active immune response were shown in the right ventricles of rhesus monkeys by the up-regulation of inflammation- and immune-related RNAs. Furthermore, peripheral blood may mediate signaling transmission among tissues to coordinate the molecular changes in the infected individuals. Collectively, these data provide insights into the pathogenesis of COVID-19 at the early stage of infection by the original SARS-CoV-2 strain and its VOCs.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Animais , SARS-CoV-2/genética , Macaca mulatta , COVID-19/genética , Perfilação da Expressão Gênica
19.
J Med Virol ; 95(6): e28846, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37282766

RESUMO

Since the first SARS-CoV-2 outbreak in late 2019, the SARS-CoV-2 genome has harbored multiple mutations, especially spike protein mutations. The currently fast-spreading Omicron variant that manifests without symptoms or with upper respiratory diseases has been recognized as a serious global public health problem. However, its pathological mechanism is largely unknown. In this work, rhesus macaques, hamsters, and BALB/C mice were employed as animal models to explore the pathogenesis of Omicron (B.1.1.529). Notably, Omicron (B.1.1.529) infected the nasal turbinates, tracheae, bronchi, and lungs of hamsters and BALB/C mice with higher viral loads than in those of rhesus macaques. Severe histopathological damage and inflammatory responses were observed in the lungs of Omicron (B.1.1.529)-infected animals. In addition, viral replication was found in multiple extrapulmonary organs. Results indicated that hamsters and BALB/c mice are potential animal models for studies on the development of drugs/vaccines and therapies for Omicron (B.1.1.529).


Assuntos
COVID-19 , SARS-CoV-2 , Camundongos , Animais , Cricetinae , Macaca mulatta , Camundongos Endogâmicos BALB C , Brônquios
20.
Gene ; 874: 147476, 2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37187243

RESUMO

Glioma is an incurable brain tumor with high recurrence due to the frequent invasion of neoplastic cells. Glucose-6-phosphate dehydrogenase (G6PD) is a critical enzyme in the pentose phosphate pathway (PPP) whose aberrant expression drives the pathogenesis of various cancers. Recent research has identified other moonlight modes of enzymes besides the well-known regulation of metabolic reprogramming. Here, we identified previously unexplored roles of G6PD in glioma via gene set variation analysis (GSVA) based on the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) database. Furthermore, survival analyses revealed that glioma patients with high G6PD expression had a worse outcome than patients with low G6PD expression (Hazard Ratio (95%CI): 2.96 (2.41, 3.64), p = 3.5E-22). Combined with functional assays, G6PD was shown to be related with the migration and invasion in glioma. G6PD knockdown could inhibit the migration in LN229 cells. And G6PD overexpression enhanced LN229 cell migration and invasion. Mechanically, the knockdown of G6PD reduced sequestosome 1 (SQSTM1) protein stability under cycloheximide (CHX) treatment. Moreover, the overexpression of SQSTM1 rescued the impaired migrated and invasive phenotypes in G6PD-silenced cells. Clinically, we validated the role of G6PD-SQSTM1 axis in glioma prognosis by constructing the multivariate cox proportional hazards regression model. These results define a pivotal function of G6PD in modulating SQSTM1 to promote glioma aggressiveness. And G6PD may be a prognostic biomarker and potential therapeutic target in glioma. G6PD-SQSTM1 axis may be a potential prognostic biomarker in glioma.


Assuntos
Glioma , Glucosefosfato Desidrogenase , Humanos , Glucosefosfato Desidrogenase/genética , Proteína Sequestossoma-1/genética , Linhagem Celular Tumoral , Glioma/patologia , Estabilidade Proteica , Biomarcadores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA