Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
JCI Insight ; 9(9)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38716725

RESUMEN

IgA nephropathy (IgAN) represents the main cause of renal failure, while the precise pathogenetic mechanisms have not been fully determined. Herein, we conducted a cross-species single-cell survey on human IgAN and mouse and rat IgAN models to explore the pathogenic programs. Cross-species single-cell RNA sequencing (scRNA-Seq) revealed that the IgAN mesangial cells (MCs) expressed high levels of inflammatory signatures CXCL12, CCL2, CSF1, and IL-34 and specifically interacted with IgAN macrophages via the CXCL12/CXCR4, CSF1/IL-34/CSF1 receptor, and integrin subunit alpha X/integrin subunit alpha M/complement C3 (C3) axes. IgAN macrophages expressed high levels of CXCR4, PDGFB, triggering receptor expressed on myeloid cells 2, TNF, and C3, and the trajectory analysis suggested that these cells derived from the differentiation of infiltrating blood monocytes. Additionally, protein profiling of 21 progression and 28 nonprogression IgAN samples revealed that proteins CXCL12, C3, mannose receptor C-type 1, and CD163 were negatively correlated with estimated glomerular filtration rate (eGFR) value and poor prognosis (30% eGFR as composite end point). Last, a functional experiment revealed that specific blockade of the Cxcl12/Cxcr4 pathway substantially attenuated the glomerulus and tubule inflammatory injury, fibrosis, and renal function decline in the mouse IgAN model. This study provides insights into IgAN progression and may aid in the refinement of IgAN diagnosis and the optimization of treatment strategies.


Asunto(s)
Progresión de la Enfermedad , Glomerulonefritis por IGA , Macrófagos , Análisis de la Célula Individual , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Ratas , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/patología , Interleucinas , Macrófagos/inmunología , Macrófagos/metabolismo , Células Mesangiales/patología , Células Mesangiales/metabolismo , Células Mesangiales/inmunología , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Ratas Wistar
2.
Eur J Radiol ; 176: 111496, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38733705

RESUMEN

PURPOSE: To develop a deep learning (DL) model for classifying histological types of primary bone tumors (PBTs) using radiographs and evaluate its clinical utility in assisting radiologists. METHODS: This retrospective study included 878 patients with pathologically confirmed PBTs from two centers (638, 77, 80, and 83 for the training, validation, internal test, and external test sets, respectively). We classified PBTs into five categories by histological types: chondrogenic tumors, osteogenic tumors, osteoclastic giant cell-rich tumors, other mesenchymal tumors of bone, or other histological types of PBTs. A DL model combining radiographs and clinical features based on the EfficientNet-B3 was developed for five-category classification. The area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity were calculated to evaluate model performance. The clinical utility of the model was evaluated in an observer study with four radiologists. RESULTS: The combined model achieved a macro average AUC of 0.904/0.873, with an accuracy of 67.5 %/68.7 %, a macro average sensitivity of 66.9 %/57.2 %, and a macro average specificity of 92.1 %/91.6 % on the internal/external test set, respectively. Model-assisted analysis improved accuracy, interpretation time, and confidence for junior (50.6 % vs. 72.3 %, 53.07[s] vs. 18.55[s] and 3.10 vs. 3.73 on a 5-point Likert scale [P < 0.05 for each], respectively) and senior radiologists (68.7 % vs. 75.3 %, 32.50[s] vs. 21.42[s] and 4.19 vs. 4.37 [P < 0.05 for each], respectively). CONCLUSION: The combined DL model effectively classified histological types of PBTs and assisted radiologists in achieving better classification results than their independent visual assessment.

3.
Insights Imaging ; 15(1): 93, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38530554

RESUMEN

OBJECTIVE: To develop a deep learning (DL) model for segmenting fat metaplasia (FM) on sacroiliac joint (SIJ) MRI and further develop a DL model for classifying axial spondyloarthritis (axSpA) and non-axSpA. MATERIALS AND METHODS: This study retrospectively collected 706 patients with FM who underwent SIJ MRI from center 1 (462 axSpA and 186 non-axSpA) and center 2 (37 axSpA and 21 non-axSpA). Patients from center 1 were divided into the training, validation, and internal test sets (n = 455, 64, and 129). Patients from center 2 were used as the external test set. We developed a UNet-based model to segment FM. Based on segmentation results, a classification model was built to distinguish axSpA and non-axSpA. Dice Similarity Coefficients (DSC) and area under the curve (AUC) were used for model evaluation. Radiologists' performance without and with model assistance was compared to assess the clinical utility of the models. RESULTS: Our segmentation model achieved satisfactory DSC of 81.86% ± 1.55% and 85.44% ± 6.09% on the internal cross-validation and external test sets. The classification model yielded AUCs of 0.876 (95% CI: 0.811-0.942) and 0.799 (95% CI: 0.696-0.902) on the internal and external test sets, respectively. With model assistance, segmentation performance was improved for the radiological resident (DSC, 75.70% vs. 82.87%, p < 0.05) and expert radiologist (DSC, 85.03% vs. 85.74%, p > 0.05). CONCLUSIONS: DL is a novel method for automatic and accurate segmentation of FM on SIJ MRI and can effectively increase radiologist's performance, which might assist in improving diagnosis and progression of axSpA. CRITICAL RELEVANCE STATEMENT: DL models allowed automatic and accurate segmentation of FM on sacroiliac joint MRI, which might facilitate quantitative analysis of FM and have the potential to improve diagnosis and prognosis of axSpA. KEY POINTS: • Deep learning was used for automatic segmentation of fat metaplasia on MRI. • UNet-based models achieved automatic and accurate segmentation of fat metaplasia. • Automatic segmentation facilitates quantitative analysis of fat metaplasia to improve diagnosis and prognosis of axial spondyloarthritis.

4.
J Pharm Biomed Anal ; 240: 115886, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38184916

RESUMEN

The generation of an immune response in neoantigen-based products relies on antigen presentation, which is closely analyzed by bioassays for T-cell functions such as tetramer or cytokine release. Mass spectrometry (MS) has the potential to directly assess the antigen-presenting capability of antigen-presenting cells (APCs), offering advantages such as speed, multi-target analysis, robustness, and ease of transferability. However, it has not been used for quality control of these products due to challenges in sensitivity, including the number of cells and peptide diversity. In this study, we describe the development and validation of an improved targeted LC-MS/MS method with high sensitivity for characterizing antigen presentation, which could be applied in the quality control of neoantigen-based products. The parameters for the extraction were carefully optimized by different short peptides. Highly sensitive targeted triple quadrupole mass spectrometry combined with ultra-high performance liquid chromatography (UHPLC) was employed using a selective ion monitoring mode (Multiple Reaction Monitoring, MRM). Besides, we successfully implemented robust quality control peptides to ensure the reliability and consistency of this method, which proved invaluable for different APCs. With reference to the guidelines from ICH Q2 (R2), M10, as well as considering the specific attributes of the product itself, we validated the method for selectivity, specificity, sensitivity, limit of detection (LOD), recovery rate, matrix effect, repeatability, and application in dendritic cells (DCs) associated with neoantigen-based products. The validation process yields satisfactory results. Combining this approach with T cell assays will comprehensively assess cell product quality attributes from physicochemical and biological perspectives.


Asunto(s)
Presentación de Antígeno , Espectrometría de Masas en Tándem , Espectrometría de Masas en Tándem/métodos , Cromatografía Liquida/métodos , Reproducibilidad de los Resultados , Cromatografía Líquida con Espectrometría de Masas , Cromatografía Líquida de Alta Presión/métodos , Péptidos
5.
Virology ; 589: 109942, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38048647

RESUMEN

Hantaan virus (HTNV) is responsible for hemorrhagic fever with renal syndrome (HFRS), primarily due to its ability to inhibit host innate immune responses, such as type I interferon (IFN-I). In this study, we conducted a transcriptome analysis to identify host factors regulated by HTNV nucleocapsid protein (NP) and glycoprotein. Our findings demonstrate that NP and Gc proteins inhibit host IFN-I production by manipulating the retinoic acid-induced gene I (RIG-I)-like receptor (RLR) pathways. Further analysis reveals that HTNV NP and Gc proteins target upstream molecules of MAVS, such as RIG-I and MDA-5, with Gc exhibiting stronger inhibition of IFN-I responses than NP. Mechanistically, NP and Gc proteins interact with tripartite motif protein 25 (TRIM25) to competitively inhibit its interaction with RIG-I/MDA5, suppressing RLR signaling pathways. Our study unveils a cross-talk between HTNV NP/Gc proteins and host immune response, providing valuable insights into the pathogenic mechanism of HTNV.


Asunto(s)
Virus Hantaan , Interferón Tipo I , Interferón Tipo I/metabolismo , Virus Hantaan/genética , Virus Hantaan/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Transducción de Señal , Inmunidad Innata , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo
6.
Infect Med (Beijing) ; 2(3): 153-166, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38073883

RESUMEN

Mpox (monkeypox) virus (MPXV), which causes a mild smallpox-like disease, has been endemic in Africa for several decades, with sporadic cases occurring in other parts of the world. However, the most recent outbreak of mpox mainly among men that have sex with men has affected several continents, posing serious global public health concerns. The infections exhibit a wide spectrum of clinical presentation, ranging from asymptomatic infection to mild, severe disease, especially in immunocompromised individuals, young children, and pregnant women. Some therapeutics and vaccines developed for smallpox have partial protective and therapeutic effects against MPXV historic isolates in animal models. However, the continued evolution of MPXV has produced multiple lineages, leading to significant gaps in the knowledge of their pathogenesis that constrain the development of targeted antiviral therapies and vaccines. MPXV infections in various animal models have provided a central platform for identification and comparison of diseased pathogenesis between the contemporary and historic isolates. In this review, we discuss the susceptibility of various animals to MPXV, and describe the key pathologic features of rodent, rabbit and nonhuman primate models. We also provide application examples of animal models in elucidating viral pathogenesis and evaluating effectiveness of vaccine and antiviral drugs. These animal models are essential to understand the biology of MPXV contemporary isolates and to rapidly test potential countermeasures. Finally, we list some remaining scientific questions of MPXV that can be resolved by animal models.

7.
Eur Radiol ; 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38127073

RESUMEN

OBJECTIVES: To develop an ensemble multi-task deep learning (DL) framework for automatic and simultaneous detection, segmentation, and classification of primary bone tumors (PBTs) and bone infections based on multi-parametric MRI from multi-center. METHODS: This retrospective study divided 749 patients with PBTs or bone infections from two hospitals into a training set (N = 557), an internal validation set (N = 139), and an external validation set (N = 53). The ensemble framework was constructed using T1-weighted image (T1WI), T2-weighted image (T2WI), and clinical characteristics for binary (PBTs/bone infections) and three-category (benign/intermediate/malignant PBTs) classification. The detection and segmentation performances were evaluated using Intersection over Union (IoU) and Dice score. The classification performance was evaluated using the receiver operating characteristic (ROC) curve and compared with radiologist interpretations. RESULT: On the external validation set, the single T1WI-based and T2WI-based multi-task models obtained IoUs of 0.71 ± 0.25/0.65 ± 0.30 for detection and Dice scores of 0.75 ± 0.26/0.70 ± 0.33 for segmentation. The framework achieved AUCs of 0.959 (95%CI, 0.955-1.000)/0.900 (95%CI, 0.773-0.100) and accuracies of 90.6% (95%CI, 79.7-95.9%)/78.3% (95%CI, 58.1-90.3%) for the binary/three-category classification. Meanwhile, for the three-category classification, the performance of the framework was superior to that of three junior radiologists (accuracy: 65.2%, 69.6%, and 69.6%, respectively) and comparable to that of two senior radiologists (accuracy: 78.3% and 78.3%). CONCLUSION: The MRI-based ensemble multi-task framework shows promising performance in automatically and simultaneously detecting, segmenting, and classifying PBTs and bone infections, which was preferable to junior radiologists. CLINICAL RELEVANCE STATEMENT: Compared with junior radiologists, the ensemble multi-task deep learning framework effectively improves differential diagnosis for patients with primary bone tumors or bone infections. This finding may help physicians make treatment decisions and enable timely treatment of patients. KEY POINTS: • The ensemble framework fusing multi-parametric MRI and clinical characteristics effectively improves the classification ability of single-modality models. • The ensemble multi-task deep learning framework performed well in detecting, segmenting, and classifying primary bone tumors and bone infections. • The ensemble framework achieves an optimal classification performance superior to junior radiologists' interpretations, assisting the clinical differential diagnosis of primary bone tumors and bone infections.

9.
J Virol ; 97(10): e0102823, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37772822

RESUMEN

IMPORTANCE: Emerging vaccine-breakthrough severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants highlight an urgent need for novel antiviral therapies. Understanding the pathogenesis of coronaviruses is critical for developing antiviral drugs. Here, we demonstrate that the SARS-CoV-2 N protein suppresses interferon (IFN) responses by reducing early growth response gene-1 (EGR1) expression. The overexpression of EGR1 inhibits SARS-CoV-2 replication by promoting IFN-regulated antiviral protein expression, which interacts with and degrades SARS-CoV-2 N protein via the E3 ubiquitin ligase MARCH8 and the cargo receptor NDP52. The MARCH8 mutants without ubiquitin ligase activity are no longer able to degrade SARS-CoV-2 N proteins, indicating that MARCH8 degrades SARS-CoV-2 N proteins dependent on its ubiquitin ligase activity. This study found a novel immune evasion mechanism of SARS-CoV-2 utilized by the N protein, which is helpful for understanding the pathogenesis of SARS-CoV-2 and guiding the design of new prevention strategies against the emerging coronaviruses.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz , Interacciones Microbiota-Huesped , SARS-CoV-2 , Ubiquitina-Proteína Ligasas , Replicación Viral , Humanos , COVID-19/virología , Descubrimiento de Drogas , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo
10.
Inflammation ; 46(6): 2402-2414, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37581761

RESUMEN

Mesangial proliferative glomerulonephritis (MsPGN), the most common pathological change in primary glomerulonephritis, is characterized by increased macrophage infiltration into glomeruli, which results in proinflammatory cytokine release. Macrophage infiltration and differentiation are induced by the Janus kinase 2 and signal transducer and activator of the transcription 1 (JAK2/STAT1) pathway. As a suppressor of cytokine signaling 1 (SOCS1) downregulates the immune response by inhibiting the JAK2/STAT1 pathway, we investigated whether a peptide mimicking the SOCS1 kinase inhibitor region, namely, SOCS1 peptidomimetic, protects against nephropathy. Glomerular JAK2/STAT1 pathway activation was synchronized with kidney injury in an MsPGN rat model. Rats treated with the SOCS1 peptidomimetic exhibited reduced pathological glomerular changes and lessened macrophage recruitment. Moreover, in vivo, the phosphorylation of the JAK2/STAT1 pathway was downregulated in infiltrated macrophages of glomeruli. In vitro, the SOCS1 peptidomimetic inhibited macrophage M1 polarization by suppressing JAK2/STAT1 activation. In conclusion, our study demonstrated that the SOCS1 peptidomimetic plays a protective role against pathologic glomerular changes in MsPGN by reducing macrophage infiltration and inhibiting macrophage polarizing to the M1 phenotype. SOCS1 peptidomimetic, therefore, presents a feasible therapeutic strategy to alleviate renal inflammation in MsPGN.


Asunto(s)
Glomerulonefritis , Peptidomiméticos , Ratas , Animales , Peptidomiméticos/farmacología , Peptidomiméticos/uso terapéutico , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Inflamación , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/patología , Citocinas/metabolismo , Macrófagos/metabolismo
11.
Biomed Pharmacother ; 165: 115233, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37536037

RESUMEN

Shenhua tablet (SH), a formulation of traditional Chinese medicine, exerts renoprotective effect on chronic kidney diseases, and it has been found to restrain inflammation, but the mechanism is still unclear. Here, we explored the potential renoprotection of SH in mesangial proliferative glomerulonephritis (MsPGN) rat model induced by anti-Thy1 antibody. Administration of SH reduced urinary albumin/creatinine ratio (UACR) and significantly attenuated mesangial cell proliferation and renal inflammation. Notably, SH protected rats against renal inflammation, which was associated with decreasing macrophage infiltration and promoting macrophage anti-inflammatory activity. Network analysis combined with arrays identified the Janus kinase signal transducer and activator of transcription (JAK-STAT) signaling pathway as the main pathways of SH could target inflammation. Furthermore, it was confirmed that mesangial cell proliferation, which response to inflammation, were alleviated by ASS1 expression enhanced after SH administration both in vivo and in vitro. Collectively, SH has the beneficial on relieving the progression of MsPGN to alleviate inflammation and mesangial proliferation by inhibiting STAT3 phosphorylation and maintains the expression level of ASS1, might be an effective strategy for treating MsPGN.


Asunto(s)
Glomerulonefritis , Nefritis , Ratas , Animales , Ratas Wistar , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Inflamación/tratamiento farmacológico , Proliferación Celular , Comprimidos/efectos adversos
12.
Emerg Infect Dis ; 29(4): 797-800, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36958012

RESUMEN

We identified Yezo virus infection in a febrile patient who had a tick bite in northeastern China, where 0.5% of Ixodes persulcatus ticks were positive for viral RNA. Clinicians should be aware of this potential health threat and include this emerging virus in the differential diagnosis for tick-bitten patients in this region.


Asunto(s)
Ixodes , Mordeduras de Garrapatas , Virosis , Virus , Animales , Humanos , China/epidemiología
13.
Adv Healthc Mater ; 12(15): e2202280, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36753620

RESUMEN

Nanomedicine exhibits emerging potentials to deliver advanced therapeutic strategies in the fight against triple-negative breast cancer (TNBC). Nevertheless, it is still difficult to develop a precise codelivery system that integrates highly effective photosensitizers, low toxicity, and hydrophobicity. In this study, PCN-224 is selected as the carrier to enable effective cancer therapy through light-activated reactive oxygen species (ROS) formation, and the PCN-224@Mn3 O4 @HA is created in a simple one-step process by coating Mn3 O4 nanoshells on the PCN-224 template, which can then be used as an "ROS activator" to exert catalase- and glutathione peroxidase-like activities to alleviate tumor hypoxia while reducing tumor reducibility, leading to improved photodynamic therapeutic (PDT) effect of PCN-224. Meanwhile, Mn2+ produced cytotoxic hydroxyl radicals (∙OH) via the Fenton-like reaction, thus producing a promising spontaneous chemodynamic therapeutic (CDT) effect. Importantly, by remodeling the tumor microenvironment (TME), Mn3 O4 nanoshells downregulated hypoxia-inducible factor 1α expression, inhibiting tumor growth and preventing tumor revival. Thus, the developed nanoshells, via light-controlled ROS formation and multimodality imaging abilities, can effectively inhibit tumor proliferation through synergistic PDT/CDT, and prevent tumor resurgence by remodeling TME.


Asunto(s)
Estructuras Metalorgánicas , Nanocáscaras , Neoplasias , Fotoquimioterapia , Humanos , Estructuras Metalorgánicas/farmacología , Especies Reactivas de Oxígeno , Línea Celular Tumoral , Microambiente Tumoral , Peróxido de Hidrógeno
14.
Cell Biosci ; 13(1): 9, 2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639652

RESUMEN

BACKGROUND: Vector-borne flaviviruses, including tick-borne encephalitis virus (TBEV), Zika virus (ZIKV), West Nile virus (WNV), yellow fever virus (YFV), dengue virus (DENV), and Japanese encephalitis virus (JEV), pose a growing threat to public health worldwide, and have evolved complex mechanisms to overcome host antiviral innate immunity. However, the underlying mechanisms of flavivirus structural proteins to evade host immune response remain elusive. RESULTS: We showed that TBEV structural protein, pre-membrane (prM) protein, could inhibit type I interferon (IFN-I) production. Mechanically, TBEV prM interacted with both MDA5 and MAVS and interfered with the formation of MDA5-MAVS complex, thereby impeding the nuclear translocation and dimerization of IRF3 to inhibit RLR antiviral signaling. ZIKV and WNV prM was also demonstrated to interact with both MDA5 and MAVS, while dengue virus serotype 2 (DENV2) and YFV prM associated only with MDA5 or MAVS to suppress IFN-I production. In contrast, JEV prM could not suppress IFN-I production. Overexpression of TBEV and ZIKV prM significantly promoted the replication of TBEV and Sendai virus. CONCLUSION: Our findings reveal the immune evasion mechanisms of flavivirus prM, which may contribute to understanding flavivirus pathogenicity, therapeutic intervention and vaccine development.

16.
Acta Pharmacol Sin ; 44(2): 381-392, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35840657

RESUMEN

Acute kidney injury (AKI) refers to a group of common clinical syndromes characterized by acute renal dysfunction, which may lead to chronic kidney disease (CKD), and this process is called the AKI-CKD transition. The transcriptional coactivator YAP can promote the AKI-CKD transition by regulating the expression of profibrotic factors, and 14-3-3 protein zeta (14-3-3ζ), an important regulatory protein of YAP, may prevent the AKI-CKD transition. We established an AKI-CKD model in mice by unilateral renal ischemia-reperfusion injury and overexpressed 14-3-3ζ in mice using a fluid dynamics-based gene transfection technique. We also overexpressed and knocked down 14-3-3ζ in vitro. In AKI-CKD model mice, 14-3-3ζ expression was significantly increased at the AKI stage. During the development of chronic disease, the expression of 14-3-3ζ tended to decrease, whereas active YAP was consistently overexpressed. In vitro, we found that 14-3-3ζ can combine with YAP, promote the phosphorylation of YAP, inhibit YAP nuclear translocation, and reduce the expression of fibrosis-related proteins. In an in vivo intervention experiment, we found that the overexpression of 14-3-3ζ slowed the process of renal fibrosis in a mouse model of AKI-CKD. These findings suggest that 14-3-3ζ can affect the expression of fibrosis-related proteins by regulating YAP, inhibit the maladaptive repair of renal tubular epithelial cells, and prevent the AKI-CKD transition.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Daño por Reperfusión , Ratones , Animales , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Riñón/patología , Insuficiencia Renal Crónica/metabolismo , Lesión Renal Aguda/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fibrosis , Daño por Reperfusión/patología
17.
PLoS Negl Trop Dis ; 16(12): e0011017, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36542659

RESUMEN

BACKGROUND: Ticks act as important vectors of infectious agents, and several emerging tick-borne viruses have recently been identified to be associated with human diseases in northeastern China. However, little is known about the tick virome in northeastern China. METHODS: Ticks collected from April 2020 to July 2021 were pooled for metagenomic analysis to investigate the virome diversity in northeastern China. RESULTS: In total, 22 RNA viruses were identified, including four each in the Nairoviridae and Phenuiviridae families, three each in the Flaviviridae, Rhabdoviridae, and Solemoviridae families, two in the Chuviridae family, and one each in the Partitiviridae, Tombusviridae families and an unclassified virus. Of these, eight viruses were of novel species, belonging to the Nairoviridae (Ji'an nairovirus and Yichun nairovirus), Phenuiviridae (Mudanjiang phlebovirus), Rhabdoviridae (Tahe rhabdovirus 1-3), Chuviridae (Yichun mivirus), and Tombusviridae (Yichun tombus-like virus) families, and five members were established human pathogens, including Alongshan virus, tick-borne encephalitis virus, Songling virus, Beiji nairovirus, and Nuomin virus. I. persulcatus ticks had significant higher number of viral species than H. japonica, H. concinna, and D. silvarum ticks. Significant differences in tick viromes were observed among Daxing'an, Xiaoxing'an and Changbai mountains. CONCLUSIONS: These findings showed an extensive diversity of RNA viruses in ticks in northeastern China, revealing potential public health threats from the emerging tick-borne viruses. Further studies are needed to explain the natural circulation and pathogenicity of these viruses.


Asunto(s)
Virus ARN , Rhabdoviridae , Garrapatas , Virus , Animales , Humanos , Metagenómica , Virus ARN/genética , Virus/genética , China , Filogenia
18.
Front Microbiol ; 13: 1000322, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36238596

RESUMEN

Alongshan virus (ALSV) in the Jingmenvirus group within the family Flaviviridae is a newly discovered tick-borne virus associated with human disease, whose genome includes four segments and encodes four structural proteins (VP1a, VP1b, VP2, VP3, and VP4) and two non-structural proteins (NSP1 and NSP2). Here, we characterized the subcellular distribution and potential function of ALSV proteins in host cells. We found that viral proteins exhibited diverse subcellular distribution in multiple tissue-deriving cells and induced various morphological changes in the endoplasmic reticulum (ER), and NSP2, VP1b, VP2, and VP4 were all co-localized in the ER. The nuclear transfer and co-localization of VP4 and calnexin (a marker protein of ER), which were independent of their interaction, were unique to HepG2 cells. Expression of NSP1 could significantly reduce mitochondria quantity by inducing mitophagy. These findings would contribute to better understanding of the pathogenesis of emerging segmented flaviviruses.

19.
Microbiol Spectr ; 10(4): e0095022, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35913166

RESUMEN

This study aimed to identify hibifolin as a sortase A (SrtA) inhibitor and to determine whether it could attenuate the virulence of methicillin-resistant Staphylococcus aureus (MRSA). We employed a fluorescence resonance energy transfer (FRET) assay to screen a library of natural molecules to identify compounds that inhibit SrtA activity. Fluorescence quenching assay and molecular docking were performed to verify the direct binding interaction between SrtA and hibifolin. The pneumonia model was established using C57BL/6J mice by MRAS nasal administration for evaluating the effect of hibifolin on the pathogenicity of MRSA. Herein, we found that hibifolin was able to inhibit SrtA activity with an IC50 of 31.20 µg/mL. Further assays showed that the capacity of adhesion of bacteria to the host cells and biofilm formation was decreased in hibifolin-treated USA300. Results obtained from fluorescence quenching assay and molecular docking indicated that hibifolin was capable of targeting SrtA protein directly. This interaction was further confirmed by the finding that the inhibition activities of hibifolin on mutant SrtA were substantially reduced after mutating the binding sites (TRP-194, ALA-104, THR-180, ARG-197, ASN-114). The in vivo study showed that hibifolin in combination with cefotaxime protected mice from USA300 infection-induced pneumonia, which was more potent than cefotaxime alone, and no significant cytotoxicity of hibifolin was observed. Taken together, we identified that hibifolin attenuated the pathogenicity of S. aureus by directly targeting SrtA, which may be utilized in the future as adjuvant therapy for S. aureus infections. IMPORTANCE We identified hibifolin as a sortase A (SrtA) inhibitor by screening the natural compounds library, which effectively inhibited the activity of SrtA with an IC50 value of 31.20 µg/mL. Hibifolin attenuated the pathogenic behavior of Staphylococcus aureus, including adhesion, invasion, and biofilm formation. Binding assays showed that hibifolin bound to SrtA protein directly. Hibifolin improved the survival of pneumonia induced by S. aureus USA300 in mice and alleviated the pathological damage. Moreover, hibifolin showed a synergistic antibacterial effect with cefotaxime in USA300-infected mice.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Neumonía , Infecciones Estafilocócicas , Aminoaciltransferasas , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cefotaxima/farmacología , Cisteína Endopeptidasas , Flavonoides , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus , Virulencia
20.
Proc Natl Acad Sci U S A ; 119(30): e2123065119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858407

RESUMEN

SARS-CoV-2, the causative agent of the COVID-19 pandemic, undergoes continuous evolution, highlighting an urgent need for development of novel antiviral therapies. Here we show a quantitative mass spectrometry-based succinylproteomics analysis of SARS-CoV-2 infection in Caco-2 cells, revealing dramatic reshape of succinylation on host and viral proteins. SARS-CoV-2 infection promotes succinylation of several key enzymes in the TCA, leading to inhibition of cellular metabolic pathways. We demonstrated that host protein succinylation is regulated by viral nonstructural protein (NSP14) through interaction with sirtuin 5 (SIRT5); overexpressed SIRT5 can effectively inhibit virus replication. We found succinylation inhibitors possess significant antiviral effects. We also found that SARS-CoV-2 nucleocapsid and membrane proteins underwent succinylation modification, which was conserved in SARS-CoV-2 and its variants. Collectively, our results uncover a regulatory mechanism of host protein posttranslational modification and cellular pathways mediated by SARS-CoV-2, which may become antiviral drug targets against COVID-19.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Interacciones Huésped-Patógeno , Terapia Molecular Dirigida , Procesamiento Proteico-Postraduccional , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/metabolismo , COVID-19/virología , Células CACO-2 , Exorribonucleasas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Sirtuinas/metabolismo , Succinatos/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA