Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Exploration (Beijing) ; 4(3): 20230090, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38939861

RESUMEN

Atherosclerosis, a chronic disease associated with metabolism, poses a significant risk to human well-being. Currently, existing treatments for atherosclerosis lack sufficient efficiency, while the utilization of surface-modified nanoparticles holds the potential to deliver highly effective therapeutic outcomes. These nanoparticles can target and bind to specific receptors that are abnormally over-expressed in atherosclerotic conditions. This paper reviews recent research (2018-present) advances in various ligand-modified nanoparticle systems targeting atherosclerosis by specifically targeting signature molecules in the hope of precise treatment at the molecular level and concludes with a discussion of the challenges and prospects in this field. The intention of this review is to inspire novel concepts for the design and advancement of targeted nanomedicines tailored specifically for the treatment of atherosclerosis.

2.
Cell Mol Immunol ; 21(5): 479-494, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38443447

RESUMEN

Apart from mediating viral entry, the function of the free HIV-1 envelope protein (gp120) has yet to be elucidated. Our group previously showed that EP2 derived from one ß-strand in gp120 can form amyloid fibrils that increase HIV-1 infectivity. Importantly, gp120 contains ~30 ß-strands. We examined whether gp120 might serve as a precursor protein for the proteolytic release of amyloidogenic fragments that form amyloid fibrils, thereby promoting viral infection. Peptide array scanning, enzyme degradation assays, and viral infection experiments in vitro confirmed that many ß-stranded peptides derived from gp120 can indeed form amyloid fibrils that increase HIV-1 infectivity. These gp120-derived amyloidogenic peptides, or GAPs, which were confirmed to form amyloid fibrils, were termed gp120-derived enhancers of viral infection (GEVIs). GEVIs specifically capture HIV-1 virions and promote their attachment to target cells, thereby increasing HIV-1 infectivity. Different GAPs can cross-interact to form heterogeneous fibrils that retain the ability to increase HIV-1 infectivity. GEVIs even suppressed the antiviral activity of a panel of antiretroviral agents. Notably, endogenous GAPs and GEVIs were found in the lymphatic fluid, lymph nodes, and cerebrospinal fluid (CSF) of AIDS patients in vivo. Overall, gp120-derived amyloid fibrils might play a crucial role in the process of HIV-1 infectivity and thus represent novel targets for anti-HIV therapeutics.


Asunto(s)
Amiloide , Proteína gp120 de Envoltorio del VIH , Infecciones por VIH , VIH-1 , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/fisiología , Humanos , Amiloide/metabolismo , Infecciones por VIH/virología , Infecciones por VIH/metabolismo , Proteínas Amiloidogénicas/metabolismo , Virión/metabolismo , Péptidos/metabolismo , Péptidos/química , Péptidos/farmacología
3.
Mar Drugs ; 21(6)2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37367651

RESUMEN

Based on the marine natural products piperafizine B, XR334, and our previously reported compound 4m, fourteen novel 3,6-diunsaturated 2,5-diketopiperazine (2,5-DKP) derivatives (1, 2, 4-6, 8-16), together with two known ones (3 and 7), were designed and synthesized as anticancer agents against the A549 and Hela cell lines. The MTT assay results showed that the derivatives 6, 8-12, and 14 had moderate to good anticancer capacities, with IC50 values ranging from 0.7 to 8.9 µM. Among them, compound 11, with naphthalen-1-ylmethylene and 2-methoxybenzylidene functions at the 3 and 6 positions of 2,5-DKP ring, respectively, displayed good inhibitory activities toward both A549 (IC50 = 1.2 µM) and Hela (IC50 = 0.7 µM) cancer cells. It could also induce apoptosis and obviously block cell cycle progression in the G2/M phases in both cells at 1.0 µM. The electron-withdrawing functions might not be favorable for the derivatives with high anticancer activities. Additionally, compared to piperafizine B and XR334, these semi-N-alkylated derivatives have high liposolubilities (>1.0 mg mL-1). Compound 11 can be further developed, aiming at the discovery of a novel anticancer candidate.


Asunto(s)
Antineoplásicos , Humanos , Estructura Molecular , Relación Estructura-Actividad , Células HeLa , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Antineoplásicos/farmacología , Diseño de Fármacos , Línea Celular Tumoral , Apoptosis
4.
J Virol ; 96(24): e0124522, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36468859

RESUMEN

The global spread of the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the continuously emerging new variants underscore an urgent need for effective therapeutics for the treatment of coronavirus disease 2019 (COVID-19). Here, we screened several FDA-approved amphiphilic drugs and determined that sertraline (SRT) exhibits potent antiviral activity against infection of SARS-CoV-2 pseudovirus (PsV) and authentic virus in vitro. It effectively inhibits SARS-CoV-2 spike (S)-mediated cell-cell fusion. SRT targets the early stage of viral entry. It can bind to the S1 subunit of the S protein, especially the receptor binding domain (RBD), thus blocking S-hACE2 interaction and interfering with the proteolysis process of S protein. SRT is also effective against infection with SARS-CoV-2 PsV variants, including the newly emerging Omicron. The combination of SRT and other antivirals exhibits a strong synergistic effect against infection of SARS-CoV-2 PsV. The antiviral activity of SRT is independent of serotonin transporter expression. Moreover, SRT effectively inhibits infection of SARS-CoV-2 PsV and alleviates the inflammation process and lung pathological alterations in transduced mice in vivo. Therefore, SRT shows promise as a treatment option for COVID-19. IMPORTANCE The study shows SRT is an effective entry inhibitor against infection of SARS-CoV-2, which is currently prevalent globally. SRT targets the S protein of SARS-CoV-2 and is effective against a panel of SARS-CoV-2 variants. It also could be used in combination to prevent SARS-CoV-2 infection. More importantly, with long history of clinical use and proven safety, SRT might be particularly suitable to treat infection of SARS-CoV-2 in the central nervous system and optimized for treatment in older people, pregnant women, and COVID-19 patients with heart complications, which are associated with severity and mortality of COVID-19.


Asunto(s)
Antivirales , COVID-19 , SARS-CoV-2 , Sertralina , Glicoproteína de la Espiga del Coronavirus , Animales , Humanos , Ratones , Antivirales/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Sertralina/farmacología , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos
5.
Biomed Pharmacother ; 156: 113946, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36411632

RESUMEN

Qingfei Paidu decoction (QFPDD) has been clinically proven to be effective in the treatment of coronavirus disease 2019 (COVID-19). However, the bioactive components and therapeutic mechanisms remain unclear. This study aimed to explore the effective components and underlying mechanisms of QFPDD in the treatment of COVID-19 by targeting the virus-host interactome and verifying the antiviral activities of its active components in vitro. Key active components and targets were identified by analysing the topological features of a compound-target-pathway-disease regulatory network of QFPDD for the treatment of COVID-19. The antiviral activity of the active components was determined by a live virus infection assay, and possible mechanisms were analysed by pseudotyped virus infection and molecular docking assays. The inhibitory effects of the components tested on the virus-induced release of IL-6, IL-1ß and CXCL-10 were detected by ELISA. Three components of QFPDD, oroxylin A, hesperetin and scutellarin, exhibited potent antiviral activities against live SARS-CoV-2 virus and HCoV-OC43 virus with IC50 values ranging from 18.68 to 63.27 µM. Oroxylin A inhibited the entry of SARS-CoV-2 pseudovirus into target cells and inhibited SARS-CoV-2 S protein-mediated cell-cell fusion by binding with the ACE2 receptor. The active components of QFPDD obviously inhibited the IL-6, IL-1ß and CXCL-10 release induced by the SARS-CoV-2 S protein. This study supports the clinical application of QFPDD and provides an effective analysis method for the in-depth study of the mechanisms of traditional Chinese medicine (TCM) in the prevention and treatment of COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Humanos , Simulación del Acoplamiento Molecular , Interleucina-6 , SARS-CoV-2 , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico
6.
Front Microbiol ; 11: 504, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425892

RESUMEN

Ebola virus (EBOV), the causative pathogen of the deadly EBOV disease (EVD), can be transmitted via sexual transmission. Seminal amyloid fibrils have been found enhancers of EBOV infection. Currently, limited preventive vaccine or therapeutic is available to block EBOV infection through sexual intercourse. In this study, we repurpose tolcapone, a US Food and Drug Administration (FDA)-approved agent for Parkinson's disease, as a potent inhibitor of seminal amyloid fibrils, among which semen-derived enhancer of viral infection (SEVI) is the best-characterized. Tolcapone binds to the amyloidogenic region of the SEVI precursor peptide (PAP248-286) and inhibits PAP248-286 aggregation by disrupting PAP248-286 oligomerization. In addition, tolcapone interacts with preformed SEVI fibrils and influences the activity of SEVI in promoting infection of pseudovirus (PsV) carrying the envelope glycoprotein (GP) of the EBOV Zaire or Sudan species (Zaire PsV and Sudan PsV, respectively). Tolcapone significantly antagonizes SEVI-mediated enhancement of both Zaire PsV and Sudan PsV binding to and subsequent internalization in HeLa cells. Of note, tolcapone is also effective in inhibiting the entry of both Zaire PsV and Sudan PsV. Tolcapone inhibits viral entry possibly through binding with critical residues in EBOV GP. Moreover, the combination of tolcapone with two small-molecule entry inhibitors, including bepridil and sertraline, exhibited synergistic anti-EBOV effects in semen. Collectively, as a bifunctional agent targeting the viral infection-enhancing amyloid and the virus itself during sexual intercourse, tolcapone can act as either a prophylactic topical agent to prevent the sexual transmission of EBOV or a therapeutic to treat EBOV infection.

7.
Viruses ; 11(9)2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31540474

RESUMEN

CXC chemokine receptor 4 (CXCR4) is a co-receptor for HIV-1 entry into target cells. Its natural ligand, the chemokine SDF-1, inhibits viral entry mediated by this receptor. However, the broad expression pattern of CXCR4 and its critical roles in various physiological and pathological processes indicate that the direct application of SDF-1 as an entry inhibitor might have severe consequences. Previously, we constructed an effective SDF-1 mutant, SDF-1/54, by deleting the α-helix of the C-terminal functional region of SDF-1. Of note, SDF-1/54 shows remarkable decreased chemotoxic ability, but maintains a similar binding affinity to CXCR4, suggesting SDF-1/54 might better serve as a CXCR4 inhibitor. Here, we found that SDF-1/54 exhibited potent antiviral activity against various X4 HIV-1 strains, including the infectious clone HIV-1 NL4-3, laboratory-adapted strain HIV-1 IIIB, clinical isolates and even drug-resistant strains. By using time-of-addition assay, non-infectious and infectious cell-cell fusion assay and CXCR4 internalization assay, we demonstrated SDF-1/54 is an HIV-1 entry inhibitor. A combination of SDF-1/54 with several antiretroviral drugs exhibited potent synergistic anti-HIV-1 activity. Moreover, SDF-1/54 was stable and its anti-HIV-1 activity was not significantly affected by the presence of seminal fluid, vaginal fluid simulant and human serum albumin. SDF-1/54 showed limited in vitro cytotoxicity to lymphocytes and vaginal epithelial cells. Based on these findings, SDF-1/54 could have a therapeutic potential as an HIV-1 entry inhibitor.


Asunto(s)
Quimiocina CXCL12/farmacología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Receptores CXCR4/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos , Línea Celular , Quimiocina CXCL12/genética , Células HEK293 , Infecciones por VIH/virología , Células HeLa , Humanos , Concentración 50 Inhibidora , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
8.
J Biol Chem ; 294(37): 13740-13754, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346035

RESUMEN

Seminal amyloid fibrils are made up of naturally occurring peptide fragments and are key targets for the development of combination microbicides or antiviral drugs. Previously, we reported that the polysulfonic compound ADS-J1 is a potential candidate microbicide that not only inhibits HIV-1 entry, but also seminal fibrils. However, the carcinogenic azo moieties in ADS-J1 preclude its clinical application. Here, we screened several ADS-J1-like analogs and found that the antiparasitic drug suramin most potently inhibited seminal amyloid fibrils. Using various biochemical methods, including Congo red staining, CD analysis, transmission EM, viral infection assays, surface plasmon resonance imaging, and molecular dynamics simulations, we investigated suramin's inhibitory effects and its putative mechanism of action. We found that by forming a multivalent interaction, suramin binds to proteolytic peptides and mature fibrils, thereby inhibiting seminal fibril formation and blocking fibril-mediated enhancement of viral infection. Of note, suramin exhibited potent anti-HIV activities, and combining suramin with several antiretroviral drugs produced synergistic effects against HIV-1 in semen. Suramin also displayed a good safety profile for vaginal application. Moreover, suramin inhibited the semen-derived enhancer of viral infection (SEVI)/semen-mediated enhancement of HIV-1 transcytosis through genital epithelial cells and the subsequent infection of target cells. Collectively, suramin has great potential for further development as a combination microbicide to reduce the spread of the AIDS pandemic by targeting both viral and host factors involved in HIV-1 sexual transmission.


Asunto(s)
Amiloide/efectos de los fármacos , Semen/efectos de los fármacos , Suramina/farmacología , Adulto , Animales , Fármacos Anti-VIH/farmacología , Antirretrovirales/farmacología , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Voluntarios Sanos , Humanos , Masculino , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Conejos , Semen/metabolismo , Suramina/metabolismo
9.
Biochem J ; 476(6): 1021-1035, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30877194

RESUMEN

Semen-derived amyloid fibrils, comprising SEVI (semen-derived enhancer of viral infection) fibrils and SEM1 fibrils, could remarkably enhance HIV-1 sexual transmission and thus are potential targets for the development of an effective microbicide. Previously, we found that ADS-J1, apart from being an HIV-1 entry inhibitor, could also potently inhibit seminal amyloid fibrillization and block fibril-mediated enhancement of viral infection. However, the remodeling effects of ADS-J1 on mature seminal fibrils were unexplored. Herein, we investigated the capacity of ADS-J1 to disassemble seminal fibrils and the potential mode of action by applying several biophysical and biochemical measurements, combined with molecular dynamic (MD) simulations. We found that ADS-J1 effectively remodeled SEVI, SEM186-107 fibrils and endogenous seminal fibrils. Unlike epigallocatechin gallate (EGCG), a universal amyloid fibril breaker, ADS-J1 disaggregated SEVI fibrils into monomeric peptides, which was independent of oxidation reaction. MD simulations revealed that ADS-J1 displayed strong binding potency to the full-length PAP248-286 via electrostatic interactions, hydrophobic interactions and hydrogen bonds. ADS-J1 might initially bind to the fibrillar surface and then occupy the amyloid core, which eventually lead to fibril disassembly. Furthermore, the binding of ADS-J1 with PAP248-286 might induce conformational changes of PAP248-286 Disassembled PAP248-286 might not be favorable to re-aggregate into fibrils. ADS-J1 also exerts abilities to remodel a panel of amyloid fibrils, including Aß1-42, hIAPP1-37 and EP2 fibrils. ADS-J1 displays promising potential to be a combination microbicide and an effective lead-product to treat amyloidogenic diseases.


Asunto(s)
Proteínas Amiloidogénicas/química , Simulación de Dinámica Molecular , Naftalenosulfonatos/química , Proteínas de Plasma Seminal/química , Triazinas/química , Proteínas Amiloidogénicas/metabolismo , Línea Celular , Infecciones por VIH/metabolismo , Infecciones por VIH/transmisión , VIH-1/metabolismo , Humanos , Proteínas de Plasma Seminal/metabolismo
10.
Nat Prod Res ; 33(10): 1467-1471, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-29258357

RESUMEN

Marine micro-organisms have been proven to be excellent sources of bioactive compounds against HIV-1. Several natural products obtained from marine-derived Aspergillus fungi were screened for their activities to inhibit HIV-1 infection. Penicillixanthone A (PXA), a natural xanthone dimer from jellyfish-derived fungus Aspergillus fumigates, displayed potent anti-HIV-1 activity by inhibiting infection against CCR5-tropic HIV-1 SF162 and CXCR4-tropic HIV-1 NL4-3, with IC50 of 0.36 and 0.26 µM, respectively. Molecular docking study was conducted to understand the possible binding mode of PXA with the CCR5/CXCR4. The results revealed that, the marine-derived PXA, as a CCR5/CXCR4 dual-coreceptor antagonist, presents a new type of potential lead product for the development of anti-HIV therapeutics.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antagonistas de los Receptores CCR5/farmacología , Receptores CCR5/metabolismo , Xantonas/farmacología , Fármacos Anti-VIH/química , Organismos Acuáticos/química , Aspergillus/química , Antagonistas de los Receptores CCR5/química , Antagonistas de los Receptores CCR5/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Simulación del Acoplamiento Molecular , Receptores CCR5/química , Receptores CXCR4/antagonistas & inhibidores , Xantonas/química , Xantonas/metabolismo
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(11): 1338-1343, 2018 Nov 30.
Artículo en Chino | MEDLINE | ID: mdl-30514682

RESUMEN

OBJECTIVE: To explore the inhibitory effect of PSB0739 on the formation of semen-derived amyloid fibrils. METHODS: PAP248-286 (440 µmol/L) was incubated with PSB0739 at different concentrations, and at different time points of incubation, aliquots were taken from each sample for Congo red staining to detect the formation of amyloid fibers. The morphology of amyloid fibrils incubated in the presence or absence of PSB0739 was visualized using transmission electron microscope. The effect of PSB0739 on amyloid fibril formation was determined using virus infection assays at different time points, and the surface charges of amyloid fibril incubated with PSB0739 were calculated using a Zeta potentiometer. The cytotoxicity of PSB0739 in Hela cells was determined using MTT assay. The antiviral effect of PSB0738 against HIV- 1 was evaluated by infection assay. RESULTS: PSB0739 inhibited SEVI fibril formation in a dose-dependent manner in vitro. At 48 h of incubation, 220 µmol/L of PSB0739 obviously inhibited the formation of amyloid fibrils in 440 µmol/L of SEVI. Transmission electron microscopy revealed that 220 µmol/L PSB0739 prevented PAP248- 286 (440 µmol/L) from forming amyloid fibrils. PSB0739 antagonized SEVI-mediated enhancement of HIV-1 infection, and 1760 µmol/L of PSB0739 completely reversed the positive charge of SEVI (P < 0.05). PSB0739 below the concentration of 62.5 µmol/L showed no obvious cytotoxicity in Hela cells in vitro (P>0.05). PSB0739 showed a direct anti-HIV activity with an IC50 of 21.77±5.15 µmol/L. CONCLUSIONS: PSB0739 can inhibit the formation of semen-derived amyloid fibrils in vitro.


Asunto(s)
Amiloide/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Semen/química , Amiloide/química , Fármacos Anti-VIH/farmacología , Relación Dosis-Respuesta a Droga , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Células HeLa , Humanos , Técnicas In Vitro , Microscopía Electrónica de Transmisión
12.
Front Microbiol ; 9: 1330, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29971062

RESUMEN

Several studies have reported that amyloid fibrils in human semen formed from a naturally occurring peptide fragment of prostatic acidic phosphatase (PAP248-286), known as semen-derived enhancer of viral infection (SEVI), could dramatically enhance human immunodeficiency virus type 1 (HIV-1) infection. Accordingly, SEVI might serve as a novel target for new antiviral drugs or microbicide candidates for the prevention of sexually transmitted HIV. Theoretically, a special anti-PAP or anti-SEVI antibody could reduce the enhancement of viral infection by blocking the binding of HIV and SEVI fibrils. Here, 3-hydroxyphthalic anhydride modified anti-PAP248-286 antibody, named HP-API, exhibited broad-spectrum and highly effective anti-HIV-1 activities on different subtypes and tropism. By using time-of-addition, cell-cell fusion and a single-cycle HIV-1 infection assays, we demonstrated that HP-API is an HIV-1 entry/fusion inhibitor. Mechanism studies suggest that HP-API inhibited HIV-1 entry/fusion by targeting both HIV-1 gp120 envelop and CD4 receptor on the host cell specifically. It is noteworthy that HP-API abrogated the formation of SEVI fibrils and partially interfered with SEVI-mediated enhancement of HIV-1 infection. Based on these findings, HP-API could be considered a bifunctional HIV-1 entry/fusion inhibitor with high potential.

13.
ChemistryOpen ; 7(6): 447-456, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29928568

RESUMEN

The peptide segment of prostatic acid phosphatase (PAP248-286) aggregates to form SEVI (semen-derived enhancer of virus infection) amyloid fibrils. These are characteristic seminal amyloids that have the ability to promote the effect of HIV infection. In this paper, we explore the binding of sulfonated compounds with PAP248-286 through an in silico study. Three derivatives of suramin, NF110, NF279, and NF340, are selected. All of these sulfonated molecules bind to PAP248-286 and alter the conformation of the peptide, even though they have various structures, sizes, and configurations. The compounds bind with PAP248-286 through multiple interactions, such as hydrogen-bonding interactions, hydrophobic interactions, π-π stacking interactions, and electrostatic interactions. However, NF110, which has an X-shaped configuration, has the highest binding affinity of the three derivatives investigated. We also perform surface plasmon resonance and a Congo red assay to validate the results. The interactions between PAP248-286 and the sulfonated compounds are proposed to depend on the orientations of the sulfonate groups and the specific configurations of the compounds instead of the number of sulfonate groups. NF110 molecules occupy the exposed binding sites of PAP248-286, blocking interactions between the peptides. Therefore, these compounds are important in inhibiting the aggregation of PAP248-286. Herein, we provide useful information to develop new efficient microbicides to antagonize seminal amyloid fibrils and to block HIV transmission.

14.
J Nat Med ; 72(3): 808-815, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29623508

RESUMEN

Two new dyhydrophaseic acid glucoside isomers, (1'S, 3'R, 5'S, 8'R, 2Z, 4E)-dihydrophaseic acid-3'-O-ß-D-glucopyranoside (2) and (1'R, 3'S, 5'R, 8'R, 2Z, 4E)-dihydrophaseic acid-3'-O-ß-D-glucopyranoside (4), together with 10 known compounds [myo-inositol (1), 3,4-dihydroxybenzoic acid (3), 3-O-galloyl quinic acid (5), ellagic acid (6), gallic acid (7), ethyl gallate (8), scopoletin (9), ellagic acid-4-O-ß-D-glucopyranoside (10), ellagic acid-4-O-α-L-rhamnopyranoside (11), and isocorilagin (12)] were isolated from the chloroform extract of Canarium album Raeusch fruits by repeated chromatography on macroporous adsorption resin, silica gel, Sephadex LH-20, Toyopearl HW-40F, and reverse-phase C18 columns, etc. Their structures and absolute configurations were determined by comprehensive analysis of 1D- and 2D-nuclear magnetic resonance (NMR), high-resolution electron spray ionization mass spectrometry (HR-ESI-MS), ESI-MS, optical rotation, circular dichroism spectra, and comparison of NMR data with data of known compounds. Bioassay of their anti-influenza virus A activities showed that compounds 9 and 12 displayed a significant inhibitory effect with IC50 values of 22.9 ± 3.7 and 5.42 ± 0.97 µg/ml, respectively.


Asunto(s)
Burseraceae/química , Frutas/química , Virus de la Influenza A/efectos de los fármacos , Extractos Vegetales/química
15.
Biophys J ; 113(7): 1425-1439, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978437

RESUMEN

Identification of the host or viral factors that enhance HIV infection is critical for preventing sexual transmission of HIV. Amyloid fibrils derived from human semen, including semen-derived enhancer of virus infection and semenogelins, enhance HIV-1 infection dramatically in vitro. In this study, we reported that a short-degraded peptide fragment 1 (DPF1) derived from native HIV-1 envelope protein gp120-loaded rat hepatocytes, formed fibrils by self-assembly and thus enhanced HIV-1 infection by promoting the binding of HIV-1 to target cells. Furthermore, DPF1-formed fibrils might be used as a crossing seed to accelerate the formation of semen-derived enhancer of virus infection and semenogelin fibrils. It will be helpful to clarify the viral factors that affect HIV-1 infection. DPF1 as an analog of gp120 containing the critical residues for CD4 binding might be useful for designing of HIV vaccines and developing HIV entry inhibitors.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/metabolismo , VIH-1 , Hepatocitos/metabolismo , Hepatocitos/virología , Amiloide/metabolismo , Animales , Línea Celular , Supervivencia Celular , Dicroismo Circular , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/ultraestructura , Hepatocitos/patología , Humanos , Microscopía de Fuerza Atómica , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteolisis , Ratas , Semen/metabolismo , Semen/virología , Virión/metabolismo
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(7): 907-913, 2017 Jul 20.
Artículo en Chino | MEDLINE | ID: mdl-28736366

RESUMEN

OBJECTIVE: To investigate the inhibitory effect of lactic acid on semen-derived amyloid (SEVI) fibril formation. METHODS: PAP248-286 (2 mg/mL) was incubated with 4.0, 2.0, 1.0, 0.5, 0.25, and 0.125 mg/mL of lactic acid. After incubation for different times, aliquots were drawn from each sample for Thioflavin T (ThT) and Congo red staining to monitor semen-derived amyloid fibril formation. The ß sheet structure formation of PAP248-286 was measured by circular dichroism spectrum, and the morphology of amyloid fibrils incubated with or without lactic acid was observed with transmission electron microscopy (TEM). The enhancing effect of amyloid fibril incubated with lactic acid at different time points was determined using virus infection assay. PAP248-286 (2 mg/mL) was incubated with dilutions of vaginal secretion from healthy women, and amyloid fibril formation was detected with ThT and Congo red staining. RESULTS: Lactic acid inhibited SEVI fibril formation in a dose-dependent manner in vitro. Lactic acid at 0.5 mg/mL completely inhibited 2 mg/mL SEVI fibril formation within 48 h. After incubation for 48 h, lactic acid at 1 mg/mL inhibited the formation of ß-sheet structure of SEVI (2 mg/mL) and completely inhibited 2 mg/mL PAP248-286 aggregation as observed with TEM. In the presence of lactic acid, PAP248-286 lost the ability to enhance virus infection. Vaginal secretion inhibited SEVI fibril formation in a dose-dependent manner, and virtually no SEVI fibril occurred after incubation of 2 mg/mL PAP248-286 with 67% vaginal secretion. CONCLUSION: Lactic acid inhibits SEVI fibril formation in vitro.

18.
Bioorg Med Chem Lett ; 26(2): 361-365, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26711143

RESUMEN

Two new 2-benzylpyridin-4-one containing metabolites, aspernigrins C (3) and D (4), together with six known compounds (1, 2, and 5-8), were isolated from the marine-derived fungus Aspergillus niger SCSIO Jcsw6F30. The structures of the new compounds were determined by NMR, MS, and optical rotation analyses. All the isolated compounds were evaluated for their inhibitory activities against infection with HIV-1 SF162 in TZM-bl cells. Malformin C (5) showed the strongest anti-HIV-1 activity with IC50 of 1.4±0.06µM (selectivity index, 11.4), meanwhile aspernigrin C (3) also exhibited potent activity with IC50 of 4.7±0.4µM (selectivity index, 7.5).


Asunto(s)
Fármacos Anti-VIH/farmacología , Aspergillus niger/química , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Niacinamida/análogos & derivados , Piridonas/farmacología , Fármacos Anti-VIH/química , Fármacos Anti-VIH/aislamiento & purificación , Línea Celular , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Niacinamida/química , Niacinamida/aislamiento & purificación , Niacinamida/farmacología , Piridonas/química , Piridonas/aislamiento & purificación
19.
PLoS One ; 10(12): e0144522, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26656730

RESUMEN

BACKGROUND: Semen is a major vehicle for HIV transmission. Prostatic acid phosphatase (PAP) fragments, such as PAP248-286, in human semen can form amyloid fibrils to enhance HIV infection. Other endogenous or exogenous factors present during sexual intercourse have also been reported to promote the formation of seminal amyloid fibrils. METHODOLOGY AND PRINCIPAL FINDINGS: Here, we demonstrated that a synthetic 15-residue peptide derived from the HIV-1 gp120 coreceptor-binding region, designated enhancing peptide 2 (EP2), can rapidly self-assemble into nanofibers. These EP2-derivated nanofibers promptly accelerated the formation of semen amyloid fibrils by PAP248-286, as shown by Thioflavin T (ThT) and Congo red assays. The amyloid fibrils presented similar morphology, assessed via transmission electron microscopy (TEM), in the presence or absence of EP2. Circular dichroism (CD) spectroscopy revealed that EP2 accelerates PAP248-286 amyloid fibril formation by promoting the structural transition of PAP248-286 from a random coil into a cross-ß-sheet. Newly formed semen amyloid fibrils effectively enhanced HIV-1 infection in TZM-bl cells and U87 cells by promoting the binding of HIV-1 virions to target cells. CONCLUSIONS AND SIGNIFICANCE: Nanofibers composed of EP2 promote the formation of PAP248-286 amyloid fibrils and enhance HIV-1 infection.


Asunto(s)
Fosfatasa Ácida/metabolismo , Amiloide/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/patología , Agregación Patológica de Proteínas/fisiopatología , Línea Celular , VIH-1/metabolismo , Humanos , Péptidos/metabolismo , Estructura Terciaria de Proteína
20.
Antimicrob Agents Chemother ; 59(9): 5123-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26055369

RESUMEN

Semen-derived enhancer of viral infection (SEVI) is composed of amyloid fibrils that can greatly enhance HIV-1 infectivity. By its cationic property, SEVI promotes viral sexual transmission by facilitating the attachment and internalization of HIV-1 to target cells. Therefore, semen-derived amyloid fibrils are potential targets for microbicide design. ADS-J1 is an anionic HIV-1 entry inhibitor. In this study, we explored an additional function of ADS-J1: inhibition of SEVI fibril formation and blockage of SEVI-mediated enhancement of viral infection. We found that ADS-J1 bound to an amyloidogenic peptide fragment (PAP248-286, comprising amino acids 248 to 286 of the enzyme prostatic acid phosphatase), thereby inhibiting peptide assembly into amyloid fibrils. In addition, ADS-J1 binds to mature amyloid fibrils and antagonizes fibril-mediated enhancement of viral infection. Unlike cellulose sulfate, a polyanion that failed in clinical trial to prevent HIV-1 sexual transmission, ADS-J1 shows no ability to facilitate fibril formation. More importantly, the combination of ADS-J1 with several antiretroviral drugs exhibited synergistic effects against HIV-1 infection in semen, with little cytotoxicity to vaginal epithelial cells. Our results suggest that ADS-J1 or a derivative may be incorporated into a combination microbicide for prevention of the sexual transmission of HIV-1.


Asunto(s)
Amiloide/metabolismo , Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Naftalenosulfonatos/uso terapéutico , Semen/química , Triazinas/uso terapéutico , Amiloide/ultraestructura , Fármacos Anti-VIH/química , Fármacos Anti-VIH/metabolismo , Humanos , Microscopía Electrónica de Transmisión , Naftalenosulfonatos/química , Naftalenosulfonatos/metabolismo , Triazinas/química , Triazinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA