Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Retrovirology ; 15(1): 78, 2018 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30558640

RESUMO

BACKGROUND: The APOBEC3 (A3) family of DNA cytosine deaminases provides an innate barrier to infection by retroviruses including HIV-1. A total of five enzymes, A3C, A3D, A3F, A3G and A3H, are degraded by the viral accessory protein Vif and expressed at high levels in CD4+ T cells, the primary reservoir for HIV-1 replication in vivo. Apart from A3C, all of these enzymes mediate restriction of Vif-deficient HIV-1. However, a rare variant of human A3C (Ile188) was shown recently to restrict Vif-deficient HIV-1 in a 293T-based single cycle infection system. The potential activity of this naturally occurring A3C variant has yet to be characterized in a T cell-based spreading infection system. Here we employ a combination of Cas9/gRNA disruption and transient and stable protein expression to assess the roles of major Ser188 and minor Ile188 A3C variants in HIV-1 restriction in T cell lines. RESULTS: Cas9-mediated mutation of endogenous A3C in the non-permissive CEM2n T cell line did not alter HIV-1 replication kinetics, and complementation with A3C-Ser188 or A3C-Ile188 was similarly aphenotypic. Stable expression of A3C-Ser188 in the permissive T cell line SupT11 also had little effect. However, stable expression of A3C-Ile188 in SupT11 cells inhibited Vif-deficient virus replication and inflicted G-to-A mutations. CONCLUSIONS: A3C-Ile188 is capable of inhibiting Vif-deficient HIV-1 replication in T cells. Although A3C is eclipsed by the dominant anti-viral activities of other A3s in non-permissive T cell lines and primary T lymphocytes, this enzyme may still be able to contribute to HIV-1 diversification in vivo. Our results highlight the functional redundancy in the human A3 family with regards to HIV-1 restriction and the need to consider naturally occurring variants.


Assuntos
Citidina Desaminase/genética , Variação Genética , HIV-1/imunologia , Proteína 9 Associada à CRISPR/genética , Células HEK293 , HIV-1/fisiologia , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Replicação Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética
2.
Nature ; 481(7381): 371-5, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22190037

RESUMO

Restriction factors, such as the retroviral complementary DNA deaminase APOBEC3G, are cellular proteins that dominantly block virus replication. The AIDS virus, human immunodeficiency virus type 1 (HIV-1), produces the accessory factor Vif, which counteracts the host's antiviral defence by hijacking a ubiquitin ligase complex, containing CUL5, ELOC, ELOB and a RING-box protein, and targeting APOBEC3G for degradation. Here we reveal, using an affinity tag/purification mass spectrometry approach, that Vif additionally recruits the transcription cofactor CBF-ß to this ubiquitin ligase complex. CBF-ß, which normally functions in concert with RUNX DNA binding proteins, allows the reconstitution of a recombinant six-protein assembly that elicits specific polyubiquitination activity with APOBEC3G, but not the related deaminase APOBEC3A. Using RNA knockdown and genetic complementation studies, we also demonstrate that CBF-ß is required for Vif-mediated degradation of APOBEC3G and therefore for preserving HIV-1 infectivity. Finally, simian immunodeficiency virus (SIV) Vif also binds to and requires CBF-ß to degrade rhesus macaque APOBEC3G, indicating functional conservation. Methods of disrupting the CBF-ß-Vif interaction might enable HIV-1 restriction and provide a supplement to current antiviral therapies that primarily target viral proteins.


Assuntos
Subunidade beta de Fator de Ligação ao Core/metabolismo , Citidina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Desaminase APOBEC-3G , Marcadores de Afinidade , Animais , Proteínas Culina/metabolismo , Técnicas de Silenciamento de Genes , Teste de Complementação Genética , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Células Jurkat , Macaca mulatta/metabolismo , Macaca mulatta/virologia , Espectrometria de Massas , Modelos Biológicos , Ligação Proteica , Proteólise , Vírus da Imunodeficiência Símia/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Replicação Viral
4.
Commun Biol ; 4(1): 1157, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34620986

RESUMO

Organ bioengineering offers a promising solution to the persistent shortage of donor organs. However, the progression of this technology toward clinical use has been hindered by the challenges of reconstituting a functional vascular network, directing the engraftment of specific functional cell types, and defining appropriate culture conditions to concurrently support the health and phenotypic stability of diverse cell lineages. We previously demonstrated the ability to functionally reendothelialize the vasculature of a clinically scaled decellularized liver scaffold with human umbilical vein endothelial cells (HUVECs) and to sustain continuous perfusion in a large animal recovery model. We now report a method for seeding and engrafting primary porcine hepatocytes into a bioengineered liver (BEL) scaffold previously reendothelialized with HUVECs. The resulting BELs were competent for albumin production, ammonia detoxification and urea synthesis, indicating the presence of a functional hepatocyte compartment. BELs additionally slowed ammonia accumulation during in vivo perfusion in a porcine model of surgically induced acute liver failure. Following explant of the graft, BEL parenchyma showed maintenance of canonical endothelial and hepatocyte markers. Taken together, these results support the feasibility of engineering a clinically scaled functional BEL and establish a platform for optimizing the seeding and engraftment of additional liver specific cells.


Assuntos
Transplante de Fígado/métodos , Engenharia Tecidual/métodos , Animais , Modelos Animais de Doenças , Hepatócitos/transplante , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Fígado/cirurgia , Falência Hepática Aguda/cirurgia , Perfusão , Sus scrofa/cirurgia
5.
Nat Biomed Eng ; 4(4): 437-445, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31611679

RESUMO

Implanted bioengineered livers have not exceeded three days of continuous perfusion. Here we show that decellularized whole porcine livers revascularized with human umbilical vein endothelial cells and implanted heterotopically into immunosuppressed pigs whose spleens had been removed can sustain perfusion for up to 15 days. We identified peak glucose consumption rate as a main predictor of the patency of the revascularized bioengineered livers (rBELs). Heterotopic implantation of rBELs into pigs in the absence of anticoagulation therapy led to sustained perfusion for three days, followed by a pronounced immune responses directed against the human endothelial cells. A 10 day steroid-based immunosuppression protocol and a splenectomy at the time of rBEL implantation reduced the immune responses and resulted in continuous perfusion of the rBELs for over two weeks. We also show that the human endothelial cells in the perfused rBELs colonize the liver sinusoids and express sinusoidal endothelial markers similar to those in normal liver tissue. Revascularized liver scaffolds that can maintain blood perfusion at physiological pressures might eventually help to overcome the chronic shortage of transplantable human livers.


Assuntos
Engenharia Biomédica/métodos , Transplante de Fígado/métodos , Perfusão/métodos , Transplante Heterotópico/métodos , Animais , Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Células Endoteliais , Glucose , Humanos , Terapia de Imunossupressão , Cinética , Fígado/imunologia , Perfusão/instrumentação , Baço , Suínos , Alicerces Teciduais , Grau de Desobstrução Vascular
7.
Antibodies (Basel) ; 8(3)2019 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-31544853

RESUMO

The DNA cytosine deaminase APOBEC3B (A3B) is normally an antiviral factor in the innate immune response. However, A3B has been implicated in cancer mutagenesis, particularly in solid tumors of the bladder, breast, cervix, head/neck, and lung. Here, we report data on the generation and characterization of a rabbit monoclonal antibody (mAb) for human A3B. One mAb, 5210-87-13, demonstrates utility in multiple applications, including ELISA, immunoblot, immunofluorescence microscopy, and immunohistochemistry. In head-to-head tests with commercial reagents, 5210-87-13 was the only rabbit monoclonal suitable for detecting native A3B and for immunohistochemical quantification of A3B in tumor tissues. This novel mAb has the potential to enable a wide range of fundamental and clinical studies on A3B in human biology and disease.

8.
Virology ; 511: 74-81, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28841445

RESUMO

The APOBEC3 DNA cytosine deaminase family comprises a fundamental arm of the innate immune response and is best known for retrovirus restriction. Several APOBEC3 enzymes restrict HIV-1 and related retroviruses by deaminating viral cDNA cytosines to uracils compromising viral genomes. Human APOBEC3B (A3B) shows strong virus restriction activities in a variety of experimental systems, and is subjected to tight post-translational regulation evidenced by cell-specific HIV-1 restriction activity and active nuclear import. Here we ask whether lysines and/or lysine post-translational modifications are required for these A3B activities. A lysine-free derivative of human A3B was constructed and shown to be indistinguishable from the wild-type enzyme in DNA cytosine deamination, HIV-1 restriction, and nuclear localization activities. However, lysine loss did render the protein resistant to degradation by SIV Vif. Taken together, we conclude that lysine side chains and modifications thereof are unlikely to be central to A3B function or regulation in human cells.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Análise Mutacional de DNA , Lisina/genética , Lisina/metabolismo , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transporte Ativo do Núcleo Celular , Citosina/metabolismo , Desaminação , Células HEK293 , HIV-1/imunologia , Humanos , Processamento de Proteína Pós-Traducional
10.
Clin Cancer Res ; 22(18): 4746-55, 2016 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-27016308

RESUMO

PURPOSE: APOBEC3 DNA cytosine deaminase family members normally defend against viruses and transposons. However, deregulated APOBEC3 activity causes mutations in cancer. Because of broad expression profiles and varying mixtures of normal and cancer cells in tumors, including immune cell infiltration, it is difficult to determine where different APOBEC3s are expressed. Here, we ask whether correlations exist between APOBEC3 expression and T-cell infiltration in high-grade serous ovarian cancer (HGSOC), and assess whether these correlations have prognostic value. EXPERIMENTAL DESIGN: Transcripts for APOBEC3G, APOBEC3B, and the T-cell markers, CD3D, CD4, CD8A, GZMB, PRF1, and RNF128 were quantified by RT-qPCR for a cohort of 354 HGSOC patients. Expression values were correlated with each other and clinical parameters. Two additional cohorts were used to extend HGSOC clinical results. Immunoimaging was used to colocalize APOBEC3G and the T-cell marker CD3. TCGA data extended expression analyses to additional cancer types. RESULTS: A surprising positive correlation was found for expression of APOBEC3G and several T cell genes in HGSOC. Immunohistochemistry and immunofluorescent imaging showed protein colocalization in tumor-infiltrating T lymphocytes. High APOBEC3G expression correlated with improved outcomes in multiple HGSOC cohorts. TCGA data analyses revealed that expression of APOBEC3D and APOBEC3H also correlates with CD3D across multiple cancer types. CONCLUSIONS: Our results identify APOBEC3G as a new candidate biomarker for tumor-infiltrating T lymphocytes and favorable prognoses for HGSOC. Our data also highlight the complexity of the tumor environment with respect to differential APOBEC family gene expression in both tumor and surrounding normal cell types. Clin Cancer Res; 22(18); 4746-55. ©2016 AACR.


Assuntos
Desaminase APOBEC-3G/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/imunologia , Expressão Gênica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Desaminase APOBEC-3G/metabolismo , Biomarcadores Tumorais , Estudos de Coortes , Cistadenocarcinoma Seroso/mortalidade , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Ativação Linfocitária , Gradação de Tumores , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Prognóstico , Modelos de Riscos Proporcionais
11.
Sci Adv ; 1(8): e1500296, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26601257

RESUMO

A diverse set of innate immune mechanisms protects cells from viral infections. The APOBEC3 family of DNA cytosine deaminases is an integral part of these defenses. For instance, APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H would have the potential to destroy HIV-1 complementary DNA replication intermediates if not for neutralization by a proteasomal degradation mechanism directed by the viral protein Vif. At the core of this complex, Vif heterodimerizes with the transcription cofactor CBF-ß, which results in fewer transcription complexes between CBF-ß and its normal RUNX partners. Recent studies have shown that the Vif/CBF-ß interaction is specific to the primate lentiviruses HIV-1 and SIV (simian immunodeficiency virus), although related nonprimate lentiviruses still require a Vif-dependent mechanism for protection from host species' APOBEC3 enzymes. We provide a molecular explanation for this evolutionary conundrum by showing that CBF-ß is required for expression of the aforementioned HIV-1-restrictive APOBEC3 gene repertoire. Knockdown and knockout studies demonstrate that CBF-ß is required for APOBEC3 mRNA expression in the nonpermissive T cell line H9 and in primary CD4(+) T lymphocytes. Complementation experiments using CBF-ß separation-of-function alleles show that the interaction with RUNX transcription factors is required for APOBEC3 transcriptional regulation. Accordingly, the infectivity of Vif-deficient HIV-1 increases in cells lacking CBF-ß, demonstrating the importance of CBF-ß/RUNX-mediated transcription in establishing the APOBEC3 antiviral state. These findings demonstrate a major layer of APOBEC3 gene regulation in lymphocytes and suggest that primate lentiviruses evolved to hijack CBF-ß in order to simultaneously suppress this potent antiviral defense system at both transcriptional and posttranslational levels.

12.
Cell Rep ; 13(9): 1781-8, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26628363

RESUMO

APOBEC3 family DNA cytosine deaminases provide overlapping defenses against pathogen infections. However, most viruses have elaborate evasion mechanisms such as the HIV-1 Vif protein, which subverts cellular CBF-ß and a polyubiquitin ligase complex to neutralize these enzymes. Despite advances in APOBEC3 and Vif biology, a full understanding of this direct host-pathogen conflict has been elusive. We combine virus adaptation and computational studies to interrogate the APOBEC3F-Vif interface and build a robust structural model. A recurring compensatory amino acid substitution from adaptation experiments provided an initial docking constraint, and microsecond molecular dynamic simulations optimized interface contacts. Virus infectivity experiments validated a long-lasting electrostatic interaction between APOBEC3F E289 and HIV-1 Vif R15. Taken together with mutagenesis results, we propose a wobble model to explain how HIV-1 Vif has evolved to bind different APOBEC3 enzymes and, more generally, how pathogens may evolve to escape innate host defenses.


Assuntos
Citosina Desaminase/metabolismo , HIV-1/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Citosina Desaminase/química , Citosina Desaminase/genética , HIV-1/genética , Humanos , Imunidade Inata , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Eletricidade Estática , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética
13.
Structure ; 21(6): 1042-50, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23685212

RESUMO

Human APOBEC3F is an antiretroviral single-strand DNA cytosine deaminase, susceptible to degradation by the HIV-1 protein Vif. In this study the crystal structure of the HIV Vif binding, catalytically active, C-terminal domain of APOBEC3F (A3F-CTD) was determined. The A3F-CTD shares structural motifs with portions of APOBEC3G-CTD, APOBEC3C, and APOBEC2. Residues identified to be critical for Vif-dependent degradation of APOBEC3F all fit within a predominantly negatively charged contiguous region on the surface of A3F-CTD. Specific sequence motifs, previously shown to play a role in Vif susceptibility and virion encapsidation, are conserved across APOBEC3s and between APOBEC3s and HIV-1 Vif. In this structure these motifs pack against each other at intermolecular interfaces, providing potential insights both into APOBEC3 oligomerization and Vif interactions.


Assuntos
Citosina Desaminase/metabolismo , HIV-1/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Catálise , Cristalografia por Raios X , Citosina Desaminase/química , Modelos Moleculares , Conformação Proteica
14.
AIDS Res Hum Retroviruses ; 28(12): 1543-51, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22725134

RESUMO

Human immunodeficiency virus type 1 (HIV-1) requires the cellular transcription factor core binding factor subunit ß (CBFß) to stabilize its viral infectivity factor (Vif) protein and neutralize the APOBEC3 restriction factors. CBFß normally heterodimerizes with the RUNX family of transcription factors, enhancing their stability and DNA-binding affinity. To test the hypothesis that Vif may act as a RUNX mimic to bind CBFß, we generated a series of CBFß mutants at the RUNX/CBFß interface and tested their ability to stabilize Vif and impact transcription at a RUNX-dependent promoter. While several CBFß amino acid substitutions disrupted promoter activity, none of these impacted the ability of CBFß to stabilize Vif or enhance degradation of APOBEC3G. A mutagenesis screen of CBFß surface residues identified a single amino acid change, F68D, that disrupted Vif binding and its ability to degrade APOBEC3G. This mutant still bound RUNX and stimulated RUNX-dependent transcription. These separation-of-function mutants demonstrate that HIV-1 Vif and the RUNX transcription factors interact with cellular CBFß on genetically distinct surfaces.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Citidina Desaminase/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Desaminase APOBEC-3G , Substituição de Aminoácidos , Subunidade beta de Fator de Ligação ao Core/genética , Humanos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA