Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 101: 129646, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38331225

RESUMO

Dengue fever is an infectious disease caused by the dengue virus (DENV), an RNA Flavivirus transmitted by the mosquitoes Aedes aegypti and Aedes albopictus widespread in tropical, subtropical and also temperate regions. Symptoms range from a simple cold to a severe, life-threatening haemorrhagic fever. According to the WHO, it affects around 390 million people per year. No antiviral treatment for DENV is available, and the Dengvaxia vaccine is only intended for people over 9 years of age who have contracted dengue one time in the past, and shows serotype-specific effectiveness. There is therefore a crying need to discover new molecules with antiviral power against flaviviruses. The present study was carried out to evaluate the anti-DENV activities and cytotoxicity of triazenes obtained by diazocopulation. Some triazenes were highly cytotoxic (16, and 25) to hepatocarcinoma Huh7 cells, whereas others displayed strong anti-DENV potential. The antiviral activity ranged from EC50 = 7.82 µM to 48.12 µM in cellulo, with a selectivity index (CC50/EC50) greater than 9 for two of the compounds (10, and 20). In conclusion, these new triazenes could serve as a lead to develop and optimize drugs against DENV.


Assuntos
Aedes , Vírus da Dengue , Dengue , Animais , Humanos , Dengue/tratamento farmacológico , Antivirais/farmacologia
2.
Plants (Basel) ; 11(24)2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36559555

RESUMO

Major threats to the human lifespan include cancer, infectious diseases, diabetes, mental degenerative conditions and also reduced agricultural productivity due to climate changes, together with new and more devastating plant diseases. From all of this, the need arises to find new biopesticides and new medicines. Plants and microorganisms are the most important sources for isolating new metabolites. Lampedusa Island host a rich contingent of endemic species and subspecies. Seven plant species spontaneously growing in Lampedusa, i.e., Atriplex halimus L. (Ap), Daucus lopadusanus Tineo (Dl), Echinops spinosus Fiori (Es) Glaucium flavum Crantz (Gf) Hypericum aegypticum L: (Ha), Periploca angustifolia Labill (Pa), and Prasium majus L. (Pm) were collected, assessed for their metabolite content, and evaluated for potential applications in agriculture and medicine. The HPLC-MS analysis of n-hexane (HE) and CH2Cl2 (MC) extracts and the residual aqueous phases (WR) showed the presence of several metabolites in both organic extracts. Crude HE and MC extracts from Dl and He significantly inhibited butyrylcholinesterase, as did WR from the extraction of Dl and Pa. HE and MC extracts showed a significant toxicity towards hepatocarcinoma Huh7, while Dl, Ha and Er HE extracts were the most potently cytotoxic to ileocecal colorectal adenocarcinoma HCT-8 cell lines. Most extracts showed antiviral activity. At the lowest concentration tested (1.56 µg/mL), Dl, Gf and Ap MC extracts inhibited betacoronavirus HCoV-OC43 infection by> 2 fold, while the n-hexane extract of Pm was the most potent. In addition, at 1.56 µg/mL, potent inhibition (>10 fold) of dengue virus was detected for Dl, Er, and Pm HE extracts, while Pa and Ap MC extracts dampened infections to undetectable levels. Regarding to phytotoxicity, MC extracts from Er, Ap and Pm were more effective in inhibiting tomato rootlet elongation; the same first two extracts also inhibited seed cress germination while its radicle elongation, due to high sensitivity, was affected by all the extracts. Es and Gf MC extracts also inhibited seed germination of Phelipanche ramosa. Thus, we have uncovered that many of these Lampedusa plants displayed promising biopesticide, antiviral, and biological properties.

3.
Molecules ; 27(17)2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-36080382

RESUMO

Amaryllidaceae alkaloids (AAs) are a structurally diverse family of alkaloids recognized for their many therapeutic properties, such as antiviral, anti-cholinesterase, and anticancer properties. Norbelladine and its derivatives, whose biological properties are poorly studied, are key intermediates required for the biosynthesis of all ~650 reported AAs. To gain insight into their therapeutic potential, we synthesized a series of O-methylated norbelladine-type alkaloids and evaluated their cytotoxic effects on two types of cancer cell lines, their antiviral effects against the dengue virus (DENV) and the human immunodeficiency virus 1 (HIV-1), and their anti-Alzheimer's disease (anti-cholinesterase and -prolyl oligopeptidase) properties. In monocytic leukemia cells, norcraugsodine was highly cytotoxic (CC50 = 27.0 µM), while norbelladine was the most cytotoxic to hepatocarcinoma cells (CC50 = 72.6 µM). HIV-1 infection was impaired only at cytotoxic concentrations of the compounds. The 3,4-dihydroxybenzaldehyde (selectivity index (SI) = 7.2), 3',4'-O-dimethylnorbelladine (SI = 4.8), 4'-O-methylnorbelladine (SI > 4.9), 3'-O-methylnorbelladine (SI > 4.5), and norcraugsodine (SI = 3.2) reduced the number of DENV-infected cells with EC50 values ranging from 24.1 to 44.9 µM. The O-methylation of norcraugsodine abolished its anti-DENV potential. Norbelladine and its O-methylated forms also displayed butyrylcholinesterase-inhibition properties (IC50 values ranging from 26.1 to 91.6 µM). Altogether, the results provided hints of the structure−activity relationship of norbelladine-type alkaloids, which is important knowledge for the development of new inhibitors of DENV and butyrylcholinesterase.


Assuntos
Alcaloides , Alcaloides de Amaryllidaceae , Amaryllidaceae , Alcaloides/química , Alcaloides/farmacologia , Amaryllidaceae/metabolismo , Alcaloides de Amaryllidaceae/química , Antivirais/farmacologia , Butirilcolinesterase , Inibidores da Colinesterase , Humanos , Tiramina/análogos & derivados
4.
Toxins (Basel) ; 14(4)2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35448871

RESUMO

Ten Amaryllidaceae alkaloids (AAs) were isolated for the first time from Pancratium maritimum collected in Calabria region, Italy. They belong to different subgroups of this family and were identified as lycorine, which is the main alkaloid, 9-O-demethyllycorine, haemanthidine, haemanthamine, 11-hydroxyvittatine, homolycorine, pancracine, obliquine, tazettine and vittatine. Haemanthidine was isolated as a scalar mixture of two 6-epimers, as already known also for other 6-hydroxycrinine alkaloids, but for the first time they were separated as 6,11-O,O'-di-p-bromobenzoyl esters. The evaluation of the cytotoxic and antiviral potentials of all isolated compounds was undertaken. Lycorine and haemanthidine showed cytotoxic activity on Hacat cells and A431 and AGS cancer cells while, pancracine exhibited selective cytotoxicity against A431 cells. We uncovered that in addition to lycorine and haemanthidine, haemanthamine and pancracine also possess antiretroviral abilities, inhibiting pseudotyped human immunodeficiency virus (HIV)−1 with EC50 of 25.3 µM and 18.5 µM respectively. Strikingly, all the AAs isolated from P. maritimum were able to impede dengue virus (DENV) replication (EC50 ranged from 0.34−73.59 µM) at low to non-cytotoxic concentrations (CC50 ranged from 6.25 µM to >100 µM). Haemanthamine (EC50 = 337 nM), pancracine (EC50 = 357 nM) and haemanthidine (EC50 = 476 nM) were the most potent anti-DENV inhibitors. Thus, this study uncovered new antiviral properties of P. maritimum isolated alkaloids, a significant finding that could lead to the development of new therapeutic strategies to fight viral infectious diseases.


Assuntos
Alcaloides , Antivirais , Alcaloides/farmacologia , Antivirais/farmacologia , Humanos , Itália , Extratos Vegetais/farmacologia
5.
Molecules ; 26(23)2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34885964

RESUMO

Amaryllidaceae plants are rich in alkaloids with biological properties. Pancratium trianthum is an Amaryllidaceae species widely used in African folk medicine to treat several diseases such as central nervous system disorders, tumors, and microbial infections, and it is used to heal wounds. The current investigation explored the biological properties of alkaloid extracts from bulbs of P. trianthum collected in the Senegalese flora. Alkaloid extracts were analyzed and identified by chromatography and mass spectrometry. Alkaloid extracts from P. trianthum displayed pleiotropic biological properties. Cytotoxic activity of the extracts was determined on hepatocarcinoma Huh7 cells and on acute monocytic leukemia THP-1 cells, while agar diffusion and microdilution assays were used to evaluate antibacterial activity. Antiviral activity was measured by infection of extract-treated cells with dengue virus (DENVGFP) and human immunodeficiency virus-1 (HIV-1GFP) reporter vectors. Cytotoxicity and viral inhibition were the most striking of P. trianthum's extract activities. Importantly, non-cytotoxic concentrations were highly effective in completely preventing DENVGFP replication and in reducing pseudotyped HIV-1GFP infection levels. Our results show that P. trianthum is a rich source of molecules for the potential discovery of new treatments against various diseases. Herein, we provide scientific evidence to rationalize the traditional uses of P. trianthum for wound treatment as an anti-dermatosis and antiseptic agent.


Assuntos
Alcaloides de Amaryllidaceae/química , Alcaloides de Amaryllidaceae/farmacologia , Amaryllidaceae/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antivirais/química , Antivirais/farmacologia , Linhagem Celular Tumoral , Dengue/tratamento farmacológico , Vírus da Dengue/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Extratos Vegetais/química , Extratos Vegetais/farmacologia
6.
Virologie (Montrouge) ; 24(6): 369-380, 2020 Dec 01.
Artigo em Francês | MEDLINE | ID: mdl-33441289

RESUMO

In this review, we summarize recent advances in the knowledge of the biological functions of human TRIM5α, a cytoplasmic protein mostly known for its antiretroviral functions. In addition to directly targeting retroviral capsid cores, an inhibitory activity called "restriction", TRIM5α senses retroviruses and activates NF-κB and AP-1 signaling pathways, resulting in the production of type I interferon (IFN-I). The antiviral state resulting from the activation of these pathways includes the upregulation of other restriction factors, and is thought to be important for the control of HIV-1 in some patients. TRIM5α also targets the protease enzyme of several tick-borne flaviviruses, a family of viruses not closely related to retroviruses. In addition to these antiviral functions, TRIM5α promotes autophagy by interacting with key actors of this pathway, such as ULK1 and p62. TRIM5α may function as a selective autophagy receptor in some conditions. Altogether, our understanding of TRIM5α shows its potential for the development of medical applications in viral diseases and beyond.


Assuntos
Antivirais , HIV-1 , Fatores de Restrição Antivirais , Capsídeo , Proteínas de Transporte/genética , HIV-1/genética , Humanos , Retroviridae , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
7.
mSphere ; 2(3)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28656178

RESUMO

The PML (promyelocytic leukemia) protein is a member of the TRIM family, a large group of proteins that show high diversity in functions but possess a common tripartite motif giving the family its name. We and others recently reported that both murine PML (mPML) and human PML (hPML) strongly restrict the early stages of infection by HIV-1 and other lentiviruses when expressed in mouse embryonic fibroblasts (MEFs). This restriction activity was found to contribute to the type I interferon (IFN-I)-mediated inhibition of HIV-1 in MEFs. Additionally, PML caused transcriptional repression of the HIV-1 promoter in MEFs. In contrast, the modulation of the early stages of HIV-1 infection of human cells by PML has been investigated by RNA interference, with unclear results. In order to conclusively determine whether PML restricts HIV-1 or not in human cells, we used the clustered regularly interspaced short palindromic repeat with Cas9 (CRISPR-Cas9) system to knock out its gene in epithelial, lymphoid, and monocytic human cell lines. Infection challenges showed that PML knockout had no effect on the permissiveness of these cells to HIV-1 infection. IFN-I treatments inhibited HIV-1 equally whether PML was expressed or not. Overexpression of individual hPML isoforms, or of mPML, in a human T cell line did not restrict HIV-1. The presence of PML was not required for the restriction of nonhuman retroviruses by TRIM5α (another human TRIM protein), and TRIM5α was inhibited by arsenic trioxide through a PML-independent mechanism. We conclude that PML is not a restriction factor for HIV-1 in human cell lines representing diverse lineages. IMPORTANCE PML is involved in innate immune mechanisms against both DNA and RNA viruses. Although the mechanism by which PML inhibits highly divergent viruses is unclear, it was recently found that it can increase the transcription of interferon-stimulated genes (ISGs). However, whether human PML inhibits HIV-1 has been debated. Here we provide unambiguous, knockout-based evidence that PML does not restrict the early postentry stages of HIV-1 infection in a variety of human cell types and does not participate in the inhibition of HIV-1 by IFN-I. Although this study does not exclude the possibility of other mechanisms by which PML may interfere with HIV-1, we nonetheless demonstrate that PML does not generally act as an HIV-1 restriction factor in human cells and that its presence is not required for IFN-I to stimulate the expression of anti-HIV-1 genes. These results contribute to uncovering the landscape of HIV-1 inhibition by ISGs in human cells.

8.
Retrovirology ; 13: 19, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27000403

RESUMO

BACKGROUND: The promyelocytic leukemia (PML) protein, a type I interferon (IFN-I)-induced gene product and a member of the tripartite motif (TRIM) family, modulates the transcriptional activity of viruses belonging to various families. Whether PML has an impact on the replication of HIV-1 has not been fully addressed, but recent studies point to its possible involvement in the restriction of HIV-1 in human cells and in the maintenance of transcriptional latency in human cell lines in which HIV-1 is constitutively repressed. We investigated further the restriction of HIV-1 and a related lentivirus, SIVmac, by PML in murine cells and in a lymphocytic human cell line. In particular, we studied the relevance of PML to IFN-I-mediated inhibition and the role of individual human isoforms. RESULTS: We demonstrate that both human PML (hPML) and murine PML (mPML) inhibit the early post-entry stages of the replication of HIV-1 and a related lentivirus, SIVmac. In addition, HIV-1 was transcriptionally silenced by mPML and by hPML isoforms I, II, IV and VI in MEFs. This PML-mediated transcriptional repression was attenuated in presence of the histone deacetylase inhibitor SAHA. In contrast, depletion of PML had no effect on HIV-1 gene expression in a human T cell line. PML was found to contribute to the inhibition of HIV-1 by IFN-I. Specifically, IFN-α and IFN-ß treatments of MEFs enhanced the PML-dependent inhibition of HIV-1 early replication stages. CONCLUSIONS: We show that PML can inhibit HIV-1 and other lentiviruses as part of the IFN-I-mediated response. The restriction takes place at two distinct steps, i.e. reverse transcription and transcription, and in an isoform-specific, cellular context-specific fashion. Our results support a model in which PML activates innate immune antilentiviral effectors. These data are relevant to the development of latency reversal-inducing pharmacological agents, since PML was previously proposed as a pharmacological target for such inhibitors. This study also has implications for the development of murine models of HIV-1.


Assuntos
HIV-1/imunologia , Interações Hospedeiro-Patógeno , Proteínas Nucleares/metabolismo , Vírus da Imunodeficiência Símia/imunologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Replicação Viral , Animais , Linhagem Celular , HIV-1/fisiologia , Humanos , Interferon Tipo I/metabolismo , Camundongos , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica , Transcrição Reversa , Análise de Sequência de DNA , Vírus da Imunodeficiência Símia/fisiologia , Transcrição Gênica
9.
J Gen Virol ; 96(Pt 4): 874-886, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25502651

RESUMO

IFN-induced restriction factors can significantly affect the replicative capacity of retroviruses in mammals. TRIM5α (tripartite motif protein 5, isoform α) is a restriction factor that acts at early stages of the virus life cycle by intercepting and destabilizing incoming retroviral cores. Sensitivity to TRIM5α maps to the N-terminal domain of the retroviral capsid proteins. In several New World and Old World monkey species, independent events of retrotransposon-mediated insertion of the cyclophilin A (CypA)-coding sequence in the trim5 gene have given rise to TRIMCyp (also called TRIM5-CypA), a hybrid protein that is active against some lentiviruses in a species-specific fashion. In particular, TRIMCyp from the owl monkey (omkTRIMCyp) very efficiently inhibits human immunodeficiency virus type 1 (HIV-1). Previously, we showed that disrupting the integrity of microtubules (MTs) and of cytoplasmic dynein complexes partially rescued replication of retroviruses, including HIV-1, from restriction mediated by TRIM5α. Here, we showed that efficient restriction of HIV-1 by omkTRIMCyp was similarly dependent on the MT network and on dynein complexes, but in a context-dependent fashion. When omkTRIMCyp was expressed in human HeLa cells, restriction was partially counteracted by pharmacological agents targeting MTs or by small interfering RNA-mediated inhibition of dynein. The same drugs (nocodazole and paclitaxel) also rescued HIV-1 from restriction in cat CRFK cells, although to a lesser extent. Strikingly, neither nocodazole, paclitaxel nor depletion of the dynein heavy chain had a significant effect on the restriction of HIV-1 in an owl monkey cell line. These results suggested the existence of cell-specific functional interactions between MTs/dynein and TRIMCyp.


Assuntos
Proteínas de Transporte/farmacologia , Ciclofilina A/farmacologia , Dineínas/antagonistas & inibidores , HIV-1/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Aotidae , Gatos , Linhagem Celular , Linhagem Celular Tumoral , Citoplasma/efeitos dos fármacos , Citoplasma/virologia , Células HEK293 , Infecções por HIV/tratamento farmacológico , Células HeLa , Humanos
10.
J Virol ; 88(10): 5661-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24600008

RESUMO

UNLABELLED: The tripartite motif (TRIM) family of proteins includes the TRIM5α antiretroviral restriction factor. TRIM5α from many Old World and some New World monkeys can restrict the human immunodeficiency virus type 1 (HIV-1), while human TRIM5α restricts N-tropic murine leukemia virus (N-MLV). TRIM5α forms highly dynamic cytoplasmic bodies (CBs) that associate with and translocate on microtubules. However, the functional involvement of microtubules or other cytoskeleton-associated factors in the viral restriction process had not been shown. Here, we demonstrate the dependency of TRIM5α-mediated restriction on microtubule-mediated transport. Pharmacological disruption of the microtubule network using nocodazole or disabling it using paclitaxel (originally named taxol) decreased the restriction of N-MLV and HIV-1 by human or simian alleles of TRIM5α, respectively. In addition, pharmacological inhibition of dynein motor complexes using erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) and small interfering RNA-mediated depletion of the dynein heavy chain (DHC) similarly decreased TRIM5α-mediated restriction. The loss in restriction resulting from either the disassembly of microtubules or the disruption of dynein motor activity was seen for both endogenous and overexpressed TRIM5α and was not due to differences in protein stability or cell viability. Both nocodazole treatment and DHC depletion interfered with the dynamics of TRIM5α CBs, increasing their size and altering their intracellular localization. In addition, nocodazole, paclitaxel, and DHC depletion were all found to increase the stability of HIV-1 cores in infected cells, providing an alternative explanation for the decreased restriction. In conclusion, association with microtubules and the translocation activity of dynein motor complexes are required to achieve efficient restriction by TRIM5α. IMPORTANCE: The primate innate cellular defenses against infection by retroviruses include a protein named TRIM5α, belonging to the family of restriction factors. TRIM5α is present in the cytoplasm, where it can intercept incoming retroviruses shortly after their entry. How TRIM5α manages to be present at the appropriate subcytoplasmic location to interact with its target is unknown. We hypothesized that TRIM5α, either as a soluble protein or a high-molecular-weight complex (the cytoplasmic body), is transported within the cytoplasm by a molecular motor called the dynein complex, which is known to interact with and move along microtubules. Our results show that destructuring microtubules or crippling their function decreased the capacity of human or simian TRIM5α to restrict their retroviral targets. Inhibiting dynein motor activity, or reducing the expression of a key component of this complex, similarly affected TRIM5α-mediated restriction. Thus, we have identified specific cytoskeleton structures involved in innate antiretroviral defenses.


Assuntos
Proteínas de Transporte/metabolismo , Dineínas/metabolismo , HIV-1/imunologia , Vírus da Leucemia Murina/imunologia , Microtúbulos/metabolismo , Animais , Fatores de Restrição Antivirais , Transporte Biológico , Linhagem Celular , Humanos , Macaca mulatta , Proteínas/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
11.
Virus Res ; 184: 30-8, 2014 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-24583231

RESUMO

TRIM5α is a type I interferon-stimulated anti-retroviral restriction factor expressed in most primates and homologous proteins are expressed in other mammals. Through its C-terminal PRYSPRY (B30.2) domain, TRIM5α binds to incoming and intact post-fusion retroviral cores in the cytoplasm. Following this direct interaction, the retroviral capsid core is destabilized and progression of the virus life cycle is interrupted. Specific recognition of its viral target by TRIM5α also triggers the induction of an antiviral state involving the activation of transcription factors NF-κB- and AP-1. In addition to PRYSPRY, several other TRIM5α domains are important for anti-retroviral function, including a RING zinc-binding motif. This domain has "E3" ubiquitin ligase activity and is involved in both the direct inhibition of incoming retroviruses and innate immune activation. A highly conserved sumoylation consensus site is present between the RING motif and the N-terminal extremity of TRIM5α. No clear role in restriction has been mapped to this sumoylation site, and no sumoylated forms of TRIM5α have been observed. Here we confirm that mutating the putatively sumoylated lysine (K10) of the Rhesus macaque TRIM5α (TRIM5αRh) to an arginine has only a small effect on restriction. However, we show that the mutation significantly decreases the TRIM5α-induced generation of free K63-linked ubiquitin chains, an intermediate in the activation of innate immunity pathways. Accordingly, K10R decreases TRIM5α-mediated activation of both NF-κB and AP-1. Concomitantly, we find that K10R causes a large increase in the levels of ubiquitylated TRIM5α. Finally, treatment with the nuclear export inhibitor leptomycin B shows that K10R enhances the nuclear localization of TRIM5αRh, while at the same time reducing its level of association with nuclear SUMO bodies. In conclusion, the TRIM5α sumoylation site appears to modulate the E3 ubiquitin ligase activities of the adjacent RING domain, promoting K63-linked ubiquitin chains at the expense of auto-ubiquitylation which is probably K48-linked. Consistently, we find this sumoylation site to be important for innate immune activation by TRIM5α. In addition, lysine 10 regulates TRIM5α nuclear shuttling and nuclear localization, which may also be related to its role in innate immunity activation.


Assuntos
Proteínas/imunologia , Proteínas/metabolismo , Retroviridae/imunologia , Substituição de Aminoácidos , Animais , Arginina/genética , Arginina/metabolismo , Lisina/genética , Lisina/metabolismo , Macaca mulatta , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/metabolismo , NF-kappa B/metabolismo , Proteínas/genética , Sumoilação , Fator de Transcrição AP-1/metabolismo , Ubiquitina-Proteína Ligases
12.
Virology ; 405(2): 414-23, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20619429

RESUMO

In human cells, endogenous TRIM5alpha strongly inhibits N-tropic strains of murine leukemia virus (N-MLV) but does not target the closely related B-MLV. We have used a shRNA-based loss-of-function screen to isolate factors other than TRIM5alpha involved in the restriction of N-MLV. In one of the isolated clones, the shRNA expressed was found to target the murine double minute-2 mRNA. Knocking down MDM2 increased N-MLV and EIAV infection of human cells by 2- to 5-fold while having little effect on B-MLV. Similarly, knocking down MDM2 in African green monkey cells diminished the restriction of both N-MLV and HIV-1. Dual knockdown experiments showed that MDM2 was involved in the restriction mediated by TRIM5alpha. Moreover, MDM2 knockdown decreased the sensitivity of N-MLV infection to treatment with MG132 and As(2)O(3), two known TRIM5alpha pharmacological inhibitors. Altogether, our data suggest that MDM2 is a general but nonessential modulator of TRIM5alpha-mediated antiretroviral functions.


Assuntos
Proteínas de Transporte/metabolismo , Vírus da Anemia Infecciosa Equina/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Interferência de RNA , Animais , Fatores de Restrição Antivirais , Trióxido de Arsênio , Arsenicais/farmacologia , Proteínas de Transporte/genética , Linhagem Celular , HIV-1/genética , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/metabolismo , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/metabolismo , Leupeptinas/farmacologia , Camundongos , Óxidos/farmacologia , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
13.
Virology ; 396(2): 272-9, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19913868

RESUMO

Retroviral DNA integration leaves behind a single-strand DNA discontinuity at each virus:host DNA junction. It has long been proposed that cellular proteins detect and repair the integrated DNA and that failure to do so might lead to apoptotic cell death, but their identity remains unknown. PIKK family members ATM, DNA-PKcs and ATR have all been proposed to be important for HIV-1 replication, but these findings turned out to be very controversial. In order to clarify their role in retroviral replication, we analyzed the effect of pharmacological inhibitors and of a dominant-negative version of ATR on the replication of retroviruses in cell lines relevant to HIV-1 infection. Our data show that ATR and probably other PIKKs as well are involved in retroviral replication in some but not all cell lines and that ATR increases the frequency of retroviral transduction by a mechanism other than the enhancement of infected cell survival.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Infecções por Retroviridae/virologia , Retroviridae/fisiologia , Replicação Viral/fisiologia , Androstadienos/farmacologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/farmacologia , Linhagem Celular , Transformação Celular Viral/efeitos dos fármacos , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , HIV-2/efeitos dos fármacos , HIV-2/fisiologia , Vírus da Leucemia Murina/efeitos dos fármacos , Vírus da Leucemia Murina/fisiologia , Leucemia Experimental/virologia , Inibidores de Proteínas Quinases/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/fisiologia , Transdução Genética , Infecções Tumorais por Vírus/virologia , Replicação Viral/efeitos dos fármacos , Wortmanina
14.
Retrovirology ; 4: 68, 2007 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-17892575

RESUMO

BACKGROUND: TRIM5alpha, which is expressed in most primates and the related TRIMCyp, which has been found in one of the New World monkey species, are antiviral proteins of the TRIM5 family that are able to intercept incoming retroviruses early after their entry into cells. The mechanism of action has been partially elucidated for TRIM5alpha, which seems to promote premature decapsidation of the restricted retroviruses. In addition, through its N-terminal RING domain, TRIM5alpha may sensitize retroviruses to proteasome-mediated degradation. TRIM5alpha-mediated restriction requires a physical interaction with the capsid protein of targeted retroviruses. It is unclear whether other cellular proteins are involved in the inhibition mediated by TRIM5alpha and TRIMCyp. A previous report suggested that the inhibition of HIV-1 by the rhesus macaque orthologue of TRIM5alpha was inefficient in the D17a canine cell line, suggesting that the cellular environment was important for the restriction mechanism. Here we investigated further the behavior of TRIM5alpha and TRIMCyp in the D17 cells. RESULTS: We found that the various TRIM5alpha orthologues studied (human, rhesus macaque, African green monkey) as well as TRIMCyp had poor antiviral activity in the D17 cells, despite seemingly normal expression levels and subcellular distribution. Restriction of both HIV-1 and the distantly related N-tropic murine leukemia virus (N-MLV) was low in D17 cells. Both TRIM5alpharh and TRIMCyp promoted early HIV-1 decapsidation in murine cells, but weak levels of restriction in D17 cells correlated with the absence of accelerated decapsidation in these cells and also correlated with normal levels of cDNA synthesis. Fv1, a murine restriction factor structurally unrelated to TRIM5alpha, was fully functional in D17 cells, showing that the loss of activity was specific to TRIM5alpha/TRIMCyp. CONCLUSION: We show that D17 cells provide a poor environment for the inhibition of retroviral replication by proteins of the TRIM5 family. Because both TRIM5alpha and TRIMCyp are poorly active in these cells, despite having quite different viral target recognition domains, we conclude that a step either upstream or downstream of target recognition is impaired. We speculate that an unknown factor required for TRIM5alpha and TRIMCyp activity is missing or inadequately expressed in D17 cells.


Assuntos
Antivirais/farmacologia , Ciclofilina A/metabolismo , HIV-1/efeitos dos fármacos , Vírus da Leucemia Murina/efeitos dos fármacos , Proteínas/farmacologia , Animais , Fármacos Anti-HIV/farmacologia , Linhagem Celular , Ciclofilina A/genética , Cães , Vírus da Leucemia Murina/patogenicidade , Ubiquitina-Proteína Ligases
15.
J Virol ; 80(6): 2855-62, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501094

RESUMO

Cyclophilin A (CypA), a cytoplasmic, human immunodeficiency virus type 1 (HIV-1) CA-binding protein, acts after virion membrane fusion with human cells to increase HIV-1 infectivity. HIV-1 CA is similarly greeted by CypA soon after entry into rhesus macaque or African green monkey cells, where, paradoxically, the interaction decreases HIV-1 infectivity by facilitating TRIM5alpha-mediated restriction. These observations conjure a model in which CA recognition by the human TRIM5alpha orthologue is precluded by CypA. Consistent with the model, selection of a human cell line for decreased restriction of the TRIM5alpha-sensitive, N-tropic murine leukemia virus (N-MLV) rendered HIV-1 transduction of these cells independent of CypA. Additionally, HIV-1 virus-like particles (VLPs) saturate N-MLV restriction activity, particularly when the CA-CypA interaction is disrupted. Here the effects of CypA and TRIM5alpha on HIV-1 restriction were examined directly. RNA interference was used to show that endogenous human TRIM5alpha does indeed restrict HIV-1, but the magnitude of this antiviral activity was not altered by disruption of the CA-CypA interaction or by elimination of CypA protein. Conversely, the stimulatory effect of CypA on HIV-1 infectivity was completely independent of human TRIM5alpha. Together with previous reports, these data suggest that CypA protects HIV-1 from an unknown antiviral activity in human cells. Additionally, target cell permissivity increased after loading with heterologous VLPs, consistent with a common saturable target that is epistatic to both TRIM5alpha and the putative CypA-regulated restriction factor.


Assuntos
Proteínas de Transporte/metabolismo , Ciclofilina A/metabolismo , HIV-1/patogenicidade , Fatores de Restrição Antivirais , Proteínas de Transporte/genética , Linhagem Celular , Ciclofilina A/genética , HIV-1/genética , HIV-1/fisiologia , Células HeLa , Humanos , Interferência de RNA , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Vírion/metabolismo , Replicação Viral
16.
J Virol ; 79(12): 7883-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15919943

RESUMO

TRIM5 is a determinant of species-specific differences in susceptibility to infection by retroviruses bearing particular capsids. Human immunodeficiency virus type 1 (HIV-1) infection is blocked by the alpha isoform of macaque TRIM5 (TRIM5alpha(rh)) or by the product of the owl monkey TRIM5-cyclophilin A gene fusion (TRIMCyp). Human TRIM5alpha potently restricts specific strains of murine leukemia virus (N-MLV) but has only a modest effect on HIV-1. The amino termini of TRIM5 orthologues are highly conserved and possess a coiled-coil domain that promotes homomultimerization. Here we show that heterologous expression of TRIM5alpha(rh) or TRIMCyp in human cells interferes with the anti-N-MLV activity of endogenous human TRIM5alpha (TRIM5alpha(hu)). Deletion of the cyclophilin domain from TRIMCyp has no effect on heteromultimerization or colocalization with TRIM5alpha(hu) but prevents interference with anti-N-MLV activity. These data demonstrate that TRIM5 orthologues form heteromultimers and indicate that C-terminal extensions alter virus recognition by multimers of these proteins.


Assuntos
Antivirais/metabolismo , Proteínas de Transporte/metabolismo , HIV-1/efeitos dos fármacos , Haplorrinos/metabolismo , Vírus da Leucemia Murina/efeitos dos fármacos , Proteínas/metabolismo , Animais , Fatores de Restrição Antivirais , Aotidae/metabolismo , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Ciclofilina A/genética , Ciclofilina A/metabolismo , Dimerização , HIV-1/patogenicidade , Humanos , Vírus da Leucemia Murina/patogenicidade , Macaca mulatta/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
17.
J Virol ; 79(2): 910-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15613319

RESUMO

The p6 domain of the human immunodeficiency virus type 1 (HIV-1) Gag polyprotein mediates virion budding from infected cells via protein-protein contacts with the class E vacuolar protein sorting factors, Tsg101 and AIP1/ALIX. Interaction with Tsg101 is strengthened by covalent attachment of monovalent ubiquitin to HIV-1 p6. To identify additional host factors that bind to HIV-1 p6, a human cDNA library was screened in the yeast two-hybrid system. HIV-1 p6 was found to interact with small ubiquitin-like modifier 1 (SUMO-1) as well as the E2 SUMO-1 transfer enzyme, Ubc9. Interaction with p6 was also detected with Daxx, a cellular protein to which SUMO-1 is sometimes covalently attached. SUMO-1 was incorporated into HIV-1 virions where it was protected within the virion membrane from digestion by exogenous protease. Of the two lysine residues in p6, lysine 27 uniquely served as a site of covalent SUMO-1 attachment. As previously reported, though, HIV-1 bearing the p6-K27R mutation replicated just like the wild type. Overproduction of SUMO-1 in HIV-1 producer cells had no apparent effect on virion release or on virion protein or RNA content. Infectivity of the resulting virions, though, was decreased, with the defect occurring after membrane fusion, at the time of viral cDNA synthesis. HIV-1 bearing the p6-K27R mutation was insensitive to SUMO-1 overexpression, suggesting that covalent attachment of SUMO-1 to p6 is detrimental to HIV-1 replication.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/fisiologia , Proteína SUMO-1/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Bases , Proteínas de Transporte/fisiologia , Linhagem Celular , Proteínas Correpressoras , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Chaperonas Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/fisiologia , Relação Estrutura-Atividade , Enzimas de Conjugação de Ubiquitina/fisiologia , Replicação Viral
18.
J Virol ; 78(21): 11739-50, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15479815

RESUMO

Human immunodeficiency virus type 1 (HIV-1) cDNA synthesis is inhibited in cells from some nonhuman primates by an activity called Lv1. Sensitivity to restriction by Lv1 maps to a region of the HIV-1 CA required for interaction with the cellular protein cyclophilin A. A similar antiviral activity in mammalian cells, Ref1, inhibits reverse transcription of murine leukemia virus (MLV), but only with viral strains bearing N-tropic CA. Disruption of the HIV-1 CA-cyclophilin A interaction inhibits Lv1 restriction in some cells and, paradoxically, seems to render HIV-1 sensitive to Ref1. Lv1 and Ref1 activities are overcome by high-titer infection and are saturable with nonreplicating, virus-like particles encoded by susceptible viruses. Two compounds that disrupt mitochondrial membrane potential, As(2)O(3) and m-Cl-CCP, reduce Ref1 activity. Here we show that these drugs, as well as a third compound with similar effects on mitochondria, PK11195, attenuate Lv1 activity in rhesus macaque and African green monkey cells. Effects of PK11195 and virus-like particles on HIV-1 infectivity in these cells were largely redundant, each associated with increased HIV-1 cDNA. Comparison of acutely infected macaque and human cells suggested that, in addition to effects on cDNA synthesis, Lv1 inhibits the accumulation of nuclear forms of HIV-1 cDNA. Disruption of the HIV-1 CA-cyclophilin A interaction caused a minimal increase in total viral cDNA but increased the proportion of viral cDNA in the nucleus. Consistent with a model in which Lv1 inhibits both synthesis and nuclear translocation of HIV-1 cDNA, complete suppression of macaque or African green monkey Lv1 was achieved by the additive effect of factors that stimulate both processes.


Assuntos
Transporte Ativo do Núcleo Celular , DNA Complementar/metabolismo , DNA Viral/metabolismo , HIV-1/genética , Replicação Viral , Animais , Chlorocebus aethiops , Ciclofilina A/metabolismo , Ciclosporina/farmacologia , Produtos do Gene gag/fisiologia , HIV-1/fisiologia , Humanos , Isoquinolinas/farmacologia , Macaca mulatta , Mitocôndrias/efeitos dos fármacos , Vírion/fisiologia
19.
J Bone Joint Surg Am ; 85(9): 1757-67, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12954835

RESUMO

BACKGROUND: The fate of transplanted chondrocytes used to elicit the repair of osteochondral defects is unknown. The objective of this study was to examine the fate and the expression of cartilage-specific genes in chondrocytes when the chondrocyte phenotype was maintained preoperatively by alginate suspension culture, the cells were labeled with enhanced green fluorescent protein, and the chondrocytes in alginate were then implanted into full-thickness osteochondral defects in rabbits. METHODS: To determine the effect of alginate on rabbit chondrocytes in vitro, cells were grown in monolayer or in alginate suspension culture, and gene expression for aggrecan, type-I collagen, and type-II collagen was analyzed by reverse transcription-polymerase chain reaction. Cells were genetically labeled with the gene for enhanced green fluorescent protein, and the effect of transfer of the gene for enhanced green fluorescent protein on chondrocyte phenotype was assessed in vitro. Chondrocytes labeled with enhanced green fluorescent protein that were embedded in alginate were implanted into osteochondral defects in rabbit knees, either immediately after creation of the defects or after the cells had been preconditioned in alginate suspension culture for two weeks. The repair tissue within the osteochondral defects was assessed at one to four weeks. Cells labeled with enhanced green fluorescent protein were quantified by confocal microscopy, and the repair tissue was examined histologically with safranin O. RESULTS: Gene expression by chondrocytes demonstrated a selective upregulation of cartilage-specific genes in alginate suspension culture. This effect was less pronounced in cells that were transduced with enhanced green fluorescent protein. Chondrocytes transplanted in vivo were detected in the repair tissue for the entire period of observation with diminishing cell density over time. At one week, the cell density of the transplanted chondrocytes was 100% of the initial density; at two and three weeks, the cell density was 70%; and, after four weeks, the cell density had decreased to 15%. Safranin-O staining of histological sections indicated cartilage-specific matrix production in vitro and in vivo. Integration of transplanted cells into the host repair tissue was not observed. The two-week period of preconditioning in alginate suspension culture had no apparent influence on the temporal fate of the cells or the histological appearance of the repair tissue. CONCLUSIONS AND CLINICAL RELEVANCE: Alginate promotes expression of cartilage-specific genes and allows delivery of chondrocytes into osteochondral defects. Transgenic chondrocytes labeled with enhanced green fluorescent protein are detectable in the defect, but they do not appear to form repair tissue and they decrease in number with time. In view of the clinical application of cell-based cartilage repair, understanding the fate of transplanted cells becomes increasingly relevant. Transgenic chondrocytes are an effective tool to study the role of transplanted chondrocytes in articular cartilage repair.


Assuntos
Alginatos , Cartilagem Articular/patologia , Transplante de Células/métodos , Condrócitos/transplante , Animais , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Colágeno , Meios de Cultura , Expressão Gênica , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Pediatr Orthop ; 23(4): 430-6, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12826937

RESUMO

In this study the authors explored the feasibility of using transduced cells for gene therapy to induce healing of osteochondral defects. Both a mouse mesenchymal cell line and mixed rabbit adherent stromal cells were transduced with either liposomal transfection or retroviral transduction using a traceable gene. Transduction efficiency was more than 95% with the retroviral construct and expression was maintained for over 6 months of passage. The liposomal transfection led to a transient expression with an efficiency of 50%. The expression of osteochondral genes was diminished but preserved after transduction in vitro. Transduced rabbit cells were transplanted into osteochondral defects in rabbit femoral condyles. Cells transplanted in vivo could be detected for 4 weeks in the repair tissue. The authors' data demonstrate that mesenchymal cells from bone marrow, stably transduced with a traceable gene product, retain the bone and cartilage phenotype and can be followed in vivo after transplantation into cartilage defects.


Assuntos
Transplante de Medula Óssea , Terapia Genética , Mesoderma/transplante , Osteocondrite Dissecante/terapia , Células Estromais/transplante , Animais , Linhagem Celular , Estudos de Viabilidade , Lipossomos , Mesoderma/citologia , Camundongos , Coelhos , Retroviridae , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/citologia , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA