Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(4): 622-632, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38454157

RESUMEN

The development of a vaccine specific to severe acute respiratory syndrome coronavirus 2 Omicron has been hampered due to its low immunogenicity. Here, using reverse mutagenesis, we found that a phenylalanine-to-serine mutation at position 375 (F375S) in the spike protein of Omicron to revert it to the sequence found in Delta and other ancestral strains significantly enhanced the immunogenicity of Omicron vaccines. Sequence FAPFFAF at position 371-377 in Omicron spike had a potent inhibitory effect on macrophage uptake of receptor-binding domain (RBD) nanoparticles or spike-pseudovirus particles containing this sequence. Omicron RBD enhanced binding to Siglec-9 on macrophages to impair phagocytosis and antigen presentation and promote immune evasion, which could be abrogated by the F375S mutation. A bivalent F375S Omicron RBD and Delta-RBD nanoparticle vaccine elicited potent and broad nAbs in mice, rabbits and rhesus macaques. Our research suggested that manipulation of the Siglec-9 pathway could be a promising approach to enhance vaccine response.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , Conejos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Macaca mulatta , Macrófagos , Nanovacunas , Fagocitosis , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico
2.
Bioeng Transl Med ; 9(2): e10630, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38435816

RESUMEN

Hypertrophic scar formation is influenced by the intricate interplay between fibroblasts and endothelial cells. In this study, we investigated this relationship using in vitro and in vivo models. Clinical observations revealed distinct morphological changes and increased vascularity at pathological scar sites. Further analysis using OCTA, immunohistochemistry, and immunofluorescence confirmed the involvement of angiogenesis in scar formation. Our indirect co-culture systems demonstrated that endothelial cells enhance the proliferation and migration of fibroblasts through the secretion of cytokines including VEGF, PDGF, bFGF, and TGF-ß. Additionally, a suspended co-culture multicellular spheroid model revealed molecular-level changes associated with extracellular matrix remodeling, cellular behaviors, inflammatory response, and pro-angiogenic activity. Furthermore, KEGG pathway analysis identified the involvement of TGF-ß, IL-17, Wnt, Notch, PI3K-Akt, and MAPK pathways in regulating fibroblasts activity. These findings underscore the critical role of fibroblasts-endothelial cells crosstalk in scar formation and provide potential targets for therapeutic intervention. Understanding the molecular mechanisms underlying this interplay holds promise for the development of innovative approaches to treat tissue injuries and diseases.

5.
Biomater Adv ; 149: 213369, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37058781

RESUMEN

3D extrusion bioprinting brings the prospect of stem cell-based therapies in regenerative medicine. These bioprinted stem cells are expected to proliferate and differentiate to form the desired organoids into 3D structures, which is critical for complex tissue construction. However, this strategy is hampered by low reproducible cell number and viability, and organoid immaturity due to incomplete differentiation of stem cells. Hence, we apply a novel extrusion-based bioprinting process with cellular aggregates (CA) bioink, in which the encapsulated cells are precultured in hydrogels to undergo aggregation. In this study, alginate-gelatin-collagen (Alg-Gel-Col) hydrogel containing mesenchymal stem cells (MSCs) were precultured for 48 h to form CA bioink and resulted in high cell viability and printing fidelity. Meanwhile, MSCs in CA bioink showed high proliferation, stemness and lipogenic differentiative potential in contrast to that in single cell (SC) bioink and hanging drop cell spheroid (HDCS) bioink, which indicated the considerable potential for complex tissue construction. In addition, the printability and efficacy of human umbilical cord MSCs (hUC-MSCs) were further confirmed the translational potential of this novel bioprinting method.


Asunto(s)
Bioimpresión , Células Madre Mesenquimatosas , Humanos , Bioimpresión/métodos , Diferenciación Celular , Proliferación Celular , Hidrogeles
6.
Bioact Mater ; 21: 340-357, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36185745

RESUMEN

The incorporation of vasculature is known to be effective in tissue or organ functional regeneration. However, a vague understanding of the interaction between epidermal appendages and their vascular niches is a foremost obstacle to obtaining sweat gland (SG)-specific vasculature units. Here, we map their precise anatomical connections and report that the interplay between SG cells (SGCs) and the surrounding vascular niche is key for glandular development and homeostasis maintenance. To replicate this interplay in vitro, we used three-dimensional (3D) bioprinting to generate reproducible SGC spheroids from differentiated adipose-derived mesenchymal stem cells (ADSCs). With dermal microvascular endothelial cells (DMECs), sacrificial templates made from poly (ε-caprolactone) (PCL) were fabricated to pattern the vascular niche. This interplay model promoted physiologically relevant vascularized glandular morphogenesis in vitro and in vivo. We identified a reciprocal regulatory mechanism for promoting SGs regeneration via contact-independent cell communication and direct cell-cell interactions between SGs and the vasculature. We envision the successful use of our approach for vascularized organ regeneration in the near future.

7.
Burns Trauma ; 10: tkac035, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35937591

RESUMEN

Background: Sweat glands (SGs) have low regenerative potential after severe burns or trauma and their regeneration or functional recovery still faces many obstacles. In practice, restoring SG function requires not only the structural integrity of the gland itself, but also its neighboring tissues, especially blood vessels. Collagen triple helix repeat containing-1 (CTHRC1) was first identified in vascular repair, and increasing reports showed a close correlation between cutaneous appendage specification, patterning and regeneration. The purpose of the present study was to clarify the role of CTHRC1 in SGs and their adjacent microvessels and find therapeutic strategies to restore SG function. Methods: The SGs and their adjacent microvascular network of Cthrc1 -/- mice were first investigated using sweat test, laser Doppler imaging, tissue clearing technique and transcriptome analysis. The effects of CTHRC1 on dermal microvascular endothelial cells (DMECs) were further explored with cell proliferation, DiI-labeled acetylated low-density lipoprotein uptake, tube formation and intercellular junction establishment assays. The effects of CTHRC1 on SG function restoration were finally confirmed by replenishing the protein into the paws of Cthrc1 -/- mice. Results: CTHRC1 is a key regulator of SG function in mice. At the tissue level, Cthrc1 deletion resulted in the disorder and reduction of the microvascular network around SGs. At the molecular level, the knockout of Cthrc1 reduced the expression of vascular development genes and functional proteins in the dermal tissues. Furthermore, CTHRC1 administration considerably enhanced SG function by inducing adjacent vascular network reconstruction. Conclusions: CTHRC1 promotes the development, morphogenesis and function execution of SGs and their neighboring vasculature. Our study provides a novel target for the restoration or regeneration of SG function in vivo.

8.
Int J Neurosci ; : 1-8, 2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-35880488

RESUMEN

Objective: Depression is a common psychological and physiological disease in the world, which seriously affects the quality of life of patients and families. Exercise is an economic and noninvasive antidepressant measure, which has been widely recognized and applied in daily life and clinical practice, and the related mechanism research has also been paid attention to. In recent years, a new research report pointed out that peripheral administration of L-lactate can reverse depression-like behavior in mice, which suggesting that the lactic acid produced during exercise may be one of the factors leading to antidepressant effect, but the detailed mechanism is not clear. Inflammation is the pathogenic factor of many diseases and a large number of experiments have proved that inflammation is also an important pathogenic factor leading to depression. The purpose of our experiment is to explore whether lactic acid has anti-inflammatory and antidepressant effects.Methods: Based on the LPS induced inflammatory model, animal behavior observation, protein extraction, Western blotting, immunofluorescence and other techniques were used in this experiment.Results: Lactic acid could inhibit the change of some important inflammatory factors, such as TNF-αIL-1ßphospho-NF-κB (p-NF-κB) and NLRP3 inflammasome complex (NLRP3/ASC/caspase-1) induced by LPS.Conclusion: Our current research suggested that lactic acid maybe exert antiinflammatory effect by inhibiting inflammatory factors.

9.
Adv Sci (Weinh) ; 9(11): e2105378, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35142444

RESUMEN

The SARS-CoV-2 Delta (B.1.617.2) strain is a variant of concern (VOC) that has become the dominant strain worldwide in 2021. Its transmission capacity is approximately twice that of the original strain, with a shorter incubation period and higher viral load during infection. Importantly, the breakthrough infections of the Delta variant have continued to emerge in the first-generation vaccine recipients. There is thus an urgent need to develop a novel vaccine with SARS-CoV-2 variants as the major target. Here, receptor binding domain (RBD)-conjugated nanoparticle vaccines targeting the Delta variant, as well as the early and Beta/Gamma strains, are developed. Under both a single-dose and a prime-boost strategy, these RBD-conjugated nanoparticle vaccines induce the abundant neutralizing antibodies (NAbs) and significantly protect hACE2 mice from infection by the authentic SARS-CoV-2 Delta strain, as well as the early and Beta strains. Furthermore, the elicitation of the robust production of broader cross-protective NAbs against almost all the notable SARS-CoV-2 variants including the Omicron variant in rhesus macaques by the third re-boost with trivalent vaccines is found. These results suggest that RBD-based monovalent or multivalent nanoparticle vaccines provide a promising second-generation vaccine strategy for SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Nanopartículas , Animales , Anticuerpos ampliamente neutralizantes , COVID-19/prevención & control , Macaca mulatta/metabolismo , Ratones , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Vacunas Conjugadas
10.
Immunity ; 53(6): 1315-1330.e9, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33275896

RESUMEN

Various vaccine strategies have been proposed in response to the global COVID-19 pandemic, each with unique strategies for eliciting immune responses. Here, we developed nanoparticle vaccines by covalently conjugating the self-assembled 24-mer ferritin to the receptor binding domain (RBD) and/or heptad repeat (HR) subunits of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) spike (S) protein. Compared to monomer vaccines, nanoparticle vaccines elicited more robust neutralizing antibodies and cellular immune responses. RBD and RBD-HR nanoparticle vaccinated hACE2 transgenic mice vaccinated with RBD and/or RBD-HR nanoparticles exhibited reduced viral load in the lungs after SARS-CoV-2 challenge. RBD-HR nanoparticle vaccines also promoted neutralizing antibodies and cellular immune responses against other coronaviruses. The nanoparticle vaccination of rhesus macaques induced neutralizing antibodies, and T and B cell responses prior to boost immunization; these responses persisted for more than three months. RBD- and HR-based nanoparticles thus present a promising vaccination approach against SARS-CoV-2 and other coronaviruses.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Ferritinas/inmunología , Helicobacter pylori/metabolismo , Proteínas Recombinantes de Fusión/inmunología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/inmunología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Proteínas Bacterianas/química , Vacunas contra la COVID-19/química , Ferritinas/química , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Pandemias , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/química , Vacunación
11.
Elife ; 82019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31397674

RESUMEN

The antiviral activity of host factor apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3G (APOBEC3G, A3G) and its degradation mediated by human immunodeficiency virus type 1 (HIV-1) Vif protein are important topics. Although accumulating evidence indicates the importance of deubiquitination enzymes (DUBs) in innate immunity, it is unknown if they participate in A3G stability. Here, we found that USP49 directly interacts with A3G and efficiently removes ubiquitin, consequently increasing A3G protein expression and significantly enhancing its anti-HIV-1 activity. Unexpectedly, A3G degradation was also mediated by a Vif- and cullin-ring-independent pathway, which was effectively counteracted by USP49. Furthermore, clinical data suggested that USP49 is correlated with A3G protein expression and hypermutations in Vif-positive proviruses, and inversely with the intact provirus ratio in the HIV-1 latent reservoir. Our studies demonstrated a mechanism to effectively stabilize A3G expression, which could comprise a target to control HIV-1 infection and eradicate the latent reservoir.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Factores Inmunológicos/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina/metabolismo , Replicación Viral , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata
12.
Eur J Med Chem ; 166: 480-501, 2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30739828

RESUMEN

The capsid assembly is an essential step for Hepatitis B Virus (HBV) life cycle and is an important target for anti-HBV drug development. In this report, we identified a hit compound with aminothiazole structure by the high throughput screening (HTS) which inhibited the interaction of HBV capsid protein within the cells. The structure hopping and SAR studies of the hit compound afforded compound 79 with potent anti-HBV replication activity and good basic drug-like properties. The working mechanism studies showed that compound 79 could bind to the similar binding site of known HBV capsid inhibitor with heteroaryldihydropyrimidine (HAP) scaffold, through similar hydrophobic interactions but with a different hydrogen bond. This compound exerted potent inhibitory effect upon HBV production, either in cell culture or in mice with no obvious acute toxicity. We propose that further development of this compound could lead to novel potent anti-HBV inhibitors that target HBV capsid assembly.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Proteínas de la Cápside/antagonistas & inhibidores , Virus de la Hepatitis B/efectos de los fármacos , Animales , Antivirales/química , Antivirales/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Técnicas de Química Sintética , Evaluación Preclínica de Medicamentos , Células HEK293 , Células Hep G2 , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Ensayos Analíticos de Alto Rendimiento , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Masculino , Ratones , Simulación del Acoplamiento Molecular , Conformación Proteica , Relación Estructura-Actividad , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
13.
Elife ; 82019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30652970

RESUMEN

Comprehensively elucidating the molecular mechanisms of human immunodeficiency virus type 1 (HIV-1) latency is a priority to achieve a functional cure. As current 'shock' agents failed to efficiently reactivate the latent reservoir, it is important to discover new targets for developing more efficient latency-reversing agents (LRAs). Here, we found that TRIM28 potently suppresses HIV-1 expression by utilizing both SUMO E3 ligase activity and epigenetic adaptor function. Through global site-specific SUMO-MS study and serial SUMOylation assays, we identified that P-TEFb catalytic subunit CDK9 is significantly SUMOylated by TRIM28 with SUMO4. The Lys44, Lys56 and Lys68 residues on CDK9 are SUMOylated by TRIM28, which inhibits CDK9 kinase activity or prevents P-TEFb assembly by directly blocking the interaction between CDK9 and Cyclin T1, subsequently inhibits viral transcription and contributes to HIV-1 latency. The manipulation of TRIM28 and its consequent SUMOylation pathway could be the target for developing LRAs.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/genética , Infecciones por VIH/genética , VIH-1/genética , Factor B de Elongación Transcripcional Positiva/genética , Proteína 28 que Contiene Motivos Tripartito/genética , Latencia del Virus/genética , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Factor B de Elongación Transcripcional Positiva/metabolismo , Interferencia de ARN , Sumoilación , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Replicación Viral/genética
14.
J Virol ; 92(20)2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068645

RESUMEN

Zika virus (ZIKV) is genetically and biologically related to other Flaviviridae family members and has disseminated to many countries. It is associated with severe consequences, including the abnormal development of the neural system in fetuses and neurological diseases in adults. Therefore, the development of anti-ZIKV drugs is of paramount importance. Screening of generic drugs revealed that several nonsteroidal anti-inflammatory drugs (NSAIDs), including aspirin, ibuprofen, naproxen, acetaminophen, and lornoxicam, potently inhibited the entry of Zika virus Env/HIV-1-pseudotyped viruses. They also significantly inhibited the replication of wild-type ZIKV both in cell lines and in primary human fetal endothelial cells. Interestingly, the NSAIDs exerted this inhibitory effect by potently reducing the expression of AXL, the entry cofactor of ZIKV. Further studies showed that the NSAIDs downregulated the prostaglandin E2/prostaglandin E receptor 2 (EP2)/cAMP/protein kinase A (PKA) signaling pathway and reduced PKA-dependent CDC37 phosphorylation and the interaction between CDC37 and HSP90, which subsequently facilitated CHIP/ubiquitination/proteasome-mediated AXL degradation. Taken together, our results highlight a new mechanism of action of antiviral agents which may assist in designing a convenient strategy for treating ZIKV-infected patients.IMPORTANCE Zika virus (ZIKV) infection, which causes congenital malformations, including microcephaly and other neurological disorders, has attracted global attention. We observed that several NSAIDs significantly inhibited ZIKV infection. Based on our observations, we propose a novel mechanism of action of antiviral compounds which involves the blockade of virus entry via degradation of the entry cofactor. Furthermore, NSAIDs can be practically used for preventing ZIKV infection in pregnant women, as certain NSAIDs, including ibuprofen and acetaminophen, are considered clinically safe.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Células Endoteliales/virología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Virus Zika/fisiología , Células A549 , Animales , Línea Celular , Chlorocebus aethiops , Regulación hacia Abajo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Proteolisis , Células Vero , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos , Infección por el Virus Zika/virología , Tirosina Quinasa del Receptor Axl
15.
Oncotarget ; 7(28): 44299-44309, 2016 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-27322423

RESUMEN

Pancreatic cancer is one of the most lethal human diseases, with an all-stage 5-year survival rate below 5%. To date, no effective and specific therapy is available for this disease. Mutations in KRAS are frequently reported in pancreatic and many other cancers; thus, KRAS is an attractive therapeutic target. Our objective was to specifically eliminate mutant KRAS and induce cell death of tumors expressing this mutant protein. We thus constructed several chimeric proteins by connecting the C-terminal domains of several adaptor proteins of E3 ubiquitin ligases such as CBL, CHIP, E6AP, and VHL, as well as VIF encoded by human immunodeficiency virus type 1 (HIV-1), to the Ras binding domain (RBD) of Raf. Although all of these chimeric proteins caused the degradation of mutant KRAS and the death of KRAS-mutant-tumor cell lines, the RBD-VIF with a protein transduction domain (PTD), named PTD-RBD-VIF, had the strongest tumor-killing effect. Intraperitoneally administered recombinant PTD-RBD-VIF potently inhibited the growth of xenografted KRAS-mutant pancreatic cancer cells. Our findings indicate that recombinant PTD-RBD-VIF, a chimeric protein with a combined cellular-viral origin, could be further developed for the treatment of various tumors harboring mutant or over-activated KRAS, especially for cases presenting with pancreatic cancer recurrence after surgery.


Asunto(s)
Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Recombinantes de Fusión/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
16.
Antiviral Res ; 122: 20-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26241003

RESUMEN

HIV-1 Vif protein is one of the most crucial accessory proteins for viral replication. It efficiently counteracts the important host restriction factor APOBEC3G (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3G, A3G) which is lethal to HIV-1 by causing G to A mutation of viral genome. Vif protein mediates degradation of APOBEC3G via the complicated protein-protein interactions of Vif, APOBEC3G, Elongin C/B and Cullin 5. The importance of Vif-APOBEC3G complex makes it a good potential target to develop new therapeutics of HIV-1. We identified a potent HIV-1 replication inhibitor (ZBMA-1, IC50 = 1.01 µM) that efficiently protected APOBEC3G protein by targeting Vif-APOBEC3G complex. The co-immunoprecipitation and docking studies indicated that compound ZBMA-1 affected the binding of Elongin C with Vif protein.


Asunto(s)
Fármacos Anti-VIH/farmacología , Citidina Desaminasa/química , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G , Animales , Fármacos Anti-VIH/síntesis química , Proteínas Cullin/metabolismo , Citidina Desaminasa/metabolismo , Descubrimiento de Drogas , Elonguina , VIH-1/fisiología , Humanos , Inmunoprecipitación , Ratones , Simulación del Acoplamiento Molecular , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...