Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 656
Filter
1.
J Mol Biol ; 435(3): 167924, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36535429

ABSTRACT

Members of the family Retroviridae are important animal and human pathogens. Being obligate parasites, their replication involves a series of steps during which the virus hijacks the cellular machinery. Additionally, many of the steps of retrovirus replication are unique among viruses, including reverse transcription, integration, and specific packaging of their genomic RNA (gRNA) as a dimer. Progress in retrovirology has helped identify several molecular mechanisms involved in each of these steps, but many are still unknown or remain controversial. This review summarizes our present understanding of the molecular mechanisms involved in various stages of retrovirus replication. Furthermore, it provides a comprehensive analysis of our current understanding of how different retroviruses package their gRNA into the assembling virions. RNA packaging in retroviruses holds a special interest because of the uniqueness of packaging a dimeric genome. Dimerization and packaging are highly regulated and interlinked events, critical for the virus to decide whether its unspliced RNA will be packaged as a "genome" or translated into proteins. Finally, some of the outstanding areas of exploration in the field of RNA packaging are highlighted, such as the role of epitranscriptomics, heterogeneity of transcript start sites, and the necessity of functional polyA sequences. An in-depth knowledge of mechanisms that interplay between viral and cellular factors during virus replication is critical in understanding not only the virus life cycle, but also its pathogenesis, and development of new antiretroviral compounds, vaccines, as well as retroviral-based vectors for human gene therapy.


Subject(s)
Life Cycle Stages , RNA, Viral , Retroviridae , Animals , Humans , Genomics , Retroviridae/growth & development , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Assembly/genetics , Virus Replication/genetics
2.
Commun Biol ; 4(1): 318, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750893

ABSTRACT

Bone morphogenetic protein (BMP) is a kind of classical multi-functional growth factor that plays a vital role in the formation and maintenance of bone, cartilage, muscle, blood vessels, and the regulation of adipogenesis and thermogenesis. However, understanding of the role of BMPs in antiviral immunity is still limited. Here we demonstrate that Bmp8a is a newly-identified positive regulator for antiviral immune responses. The bmp8a-/- zebrafish, when infected with viruses, show reduced antiviral immunity and increased viral load and mortality. We also show for the first time that Bmp8a interacts with Alk6a, which promotes the phosphorylation of Tbk1 and Irf3 through p38 MAPK pathway, and induces the production of type I interferons (IFNs) in response to viral infection. Our study uncovers a previously unrecognized role of Bmp8a in regulation of antiviral immune responses and provides a target for controlling viral infection.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Interferon Type I/metabolism , Retroviridae Infections/virology , Retroviridae/pathogenicity , Zebrafish Proteins/metabolism , Zebrafish/virology , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Gene Knockout Techniques , Host-Pathogen Interactions , Interferon Regulatory Factor-3/metabolism , Interferon Type I/immunology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Retroviridae/growth & development , Retroviridae/immunology , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Retroviridae Infections/metabolism , Signal Transduction , Viral Load , Virus Replication , Zebrafish/genetics , Zebrafish/immunology , Zebrafish/metabolism , Zebrafish Proteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Viruses ; 12(10)2020 10 20.
Article in English | MEDLINE | ID: mdl-33092109

ABSTRACT

One of the most important steps in any viral lifecycle is the production of progeny virions. For retroviruses as well as other viruses, this step is a highly organized process that occurs with exquisite spatial and temporal specificity on the cellular plasma membrane. To facilitate this process, retroviruses encode short peptide motifs, or L domains, that hijack host factors to ensure completion of this critical step. One such cellular machinery targeted by viruses is known as the Endosomal Sorting Complex Required for Transport (ESCRTs). Typically responsible for vesicular trafficking within the cell, ESCRTs are co-opted by the retroviral Gag polyprotein to assist in viral particle assembly and release of infectious virions. This review in the Viruses Special Issue "The 11th International Retroviral Nucleocapsid and Assembly Symposium", details recent findings that shed light on the molecular details of how ESCRTs and the ESCRT adaptor protein ALIX, facilitate retroviral dissemination at sites of viral assembly.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Retroviridae , Virus Assembly/physiology , Virus Release/physiology , HIV-1/metabolism , Nucleocapsid/metabolism , Retroviridae/growth & development , Retroviridae/metabolism , Ribonucleoproteins/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism
4.
Curr Issues Mol Biol ; 35: 1-16, 2020.
Article in English | MEDLINE | ID: mdl-31422930

ABSTRACT

Small ubiquitin-like modifier (SUMO)ylation is a crucial post-translational modification that controls functions of a wide collection of proteins and biological processes. Hence, given its pleiotropic role, viruses have developed many approaches to usurp SUMO conjugation to exploit the cellular host environment for their own benefit. Consistently, cancer cells also frequently impact on SUMO to force cellular transformation, underlining the importance of SUMO in health and diseases. Therefore, after a brief introduction to the multistep SUMOylation pathway, in this review we will focus our attention on several examples of strategies adopted by oncogenic viruses to hijack SUMOylation in order to promote infection, persistence and malignant transformation of host cells.


Subject(s)
Neoplasms/metabolism , Neoplasms/virology , Retroviridae/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Chromatin/genetics , Chromatin/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Hepacivirus/pathogenicity , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/pathogenicity , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/pathogenicity , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 1/pathogenicity , Humans , Merkel cell polyomavirus/genetics , Merkel cell polyomavirus/metabolism , Merkel cell polyomavirus/pathogenicity , Neoplasms/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomaviridae/pathogenicity , Retroviridae/genetics , Retroviridae/growth & development , Retroviridae/pathogenicity , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Curr Opin Virol ; 37: 58-62, 2019 08.
Article in English | MEDLINE | ID: mdl-31284078

ABSTRACT

Enteric viruses infect the mammalian gastrointestinal tract which is home to a diverse community of intestinal bacteria. Accumulating evidence suggests that certain enteric viruses utilize these bacteria to promote infection. While this is not surprising considering their proximity, multiple viruses from different viral families have been shown to bind directly to bacteria or bacterial components to aid in viral replication, pathogenesis, and transmission. These data suggest that the concept of a single virus infecting a single cell, independent of the environment, needs to be reevaluated. In this review, I will discuss the current knowledge of enteric virus-bacterial interactions and discuss the implications for viral pathogenesis and transmission.


Subject(s)
Gastrointestinal Tract/virology , Microbial Interactions , Microbiota , Viruses , Animals , Gastrointestinal Tract/microbiology , Host Microbial Interactions/immunology , Humans , Immune Evasion , Picornaviridae/growth & development , Picornaviridae/pathogenicity , Picornaviridae Infections/immunology , Picornaviridae Infections/microbiology , Picornaviridae Infections/transmission , Reoviridae/growth & development , Reoviridae/pathogenicity , Reoviridae Infections/immunology , Reoviridae Infections/microbiology , Reoviridae Infections/transmission , Retroviridae/growth & development , Retroviridae/pathogenicity , Retroviridae Infections/immunology , Retroviridae Infections/microbiology , Retroviridae Infections/transmission , Virus Diseases/immunology , Virus Diseases/microbiology , Virus Diseases/transmission , Virus Replication , Viruses/growth & development , Viruses/pathogenicity
6.
Viruses ; 10(8)2018 08 19.
Article in English | MEDLINE | ID: mdl-30126238

ABSTRACT

MicroRNAs (miRNAs) are small, non-coding RNAs that regulate gene expression at the post-transcriptional level. Through this activity, they are implicated in almost every cellular process investigated to date. Hence, it is not surprising that miRNAs play diverse roles in regulation of viral infections and antiviral responses. Diverse families of DNA and RNA viruses have been shown to take advantage of cellular miRNAs or produce virally encoded miRNAs that alter host or viral gene expression. MiRNA-mediated changes in gene expression have been demonstrated to modulate viral replication, antiviral immune responses, viral latency, and pathogenesis. Interestingly, viruses mediate both canonical and non-canonical interactions with miRNAs to downregulate specific targets or to promote viral genome stability, translation, and/or RNA accumulation. In this review, we focus on recent findings elucidating several key mechanisms employed by diverse virus families, with a focus on miRNAs at the host⁻virus interface during herpesvirus, polyomavirus, retroviruses, pestivirus, and hepacivirus infections.


Subject(s)
Gene Expression Regulation, Viral , Genome, Viral , Herpesviridae/genetics , MicroRNAs/genetics , Virus Diseases/genetics , Hepacivirus/genetics , Hepacivirus/growth & development , Hepacivirus/pathogenicity , Herpesviridae/growth & development , Herpesviridae/pathogenicity , Humans , Immune Evasion/genetics , MicroRNAs/classification , MicroRNAs/immunology , Nucleic Acid Conformation , Pestivirus/genetics , Pestivirus/growth & development , Pestivirus/pathogenicity , Polyomavirus/genetics , Polyomavirus/growth & development , Polyomavirus/pathogenicity , RNA, Viral/genetics , RNA, Viral/immunology , Retroviridae/genetics , Retroviridae/growth & development , Retroviridae/pathogenicity , Signal Transduction , Virus Diseases/immunology , Virus Diseases/virology , Virus Latency/genetics , Virus Replication/genetics
7.
Cell Host Microbe ; 21(6): 661-669, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-28618265

ABSTRACT

Viral epitranscriptomics is a newly emerging field that has identified unique roles for RNA modifications in modulating life cycles of RNA viruses. Despite the observation of a handful of modified viral RNAs five decades ago, very little was known about how these modifications regulate viral life cycles, until recently. Here we review the pro- and anti-viral effects of methyl-6-adenosine in distinct viral life cycles, the role of 2' O-methyl modifications in RNA stability and innate immune sensing, and functions of adenosine to inosine modifications in retroviral life cycles. With roles for over 100 modifications in RNA still unknown, this is a rapidly emerging field that is destined to suggest novel antiviral therapies.


Subject(s)
Adenosine/physiology , Life Cycle Stages , RNA Viruses/genetics , Virus Replication/genetics , Animals , Flavivirus/growth & development , Flavivirus/physiology , Humans , Immunity, Innate , Inosine/metabolism , RNA Editing/genetics , RNA Viruses/growth & development , RNA Viruses/immunology , RNA Viruses/physiology , RNA, Viral/metabolism , Retroviridae/growth & development , Retroviridae/physiology
8.
Cell Microbiol ; 19(9)2017 09.
Article in English | MEDLINE | ID: mdl-28643392

ABSTRACT

Retroviruses are obligate intracellular parasites of eukaryotic cells. After reverse transcription, the viral DNA contained in the preintegration complex is delivered to the nucleus of the host cell, where it integrates. Before reaching the nucleus, the incoming particle and the preintegration complex must travel throughout the cytoplasm. Likewise, the newly synthesized viral proteins and viral particles must transit the cytoplasm during exit. The cytoplasm is a crowded environment, and simple diffusion is difficult. Therefore, viruses have evolved to utilize the cellular mechanisms of movement through the cytoplasm, where microtubules are the roads, and the ATP-dependent motors dynein and kinesin are the vehicles for retrograde and anterograde trafficking. This review will focus on how different retroviruses (Mazon-Pfizer monkey virus, prototype foamy virus, bovine immunodeficiency virus, human immunodeficiency virus type 1, and murine leukemia virus) have subjugated the microtubule-associated motor proteins for viral replication. Although there have been advances in our understanding of how retroviruses move along microtubules, the strategies are different among them. Thus, a better understanding of the mechanisms used by each retrovirus to functionally subvert microtubule motor proteins will provide important clues in the design of new antiretroviral drugs that can specifically disrupt intracellular viral trafficking.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Retroviridae/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA, Viral/genetics , Humans , Protein Transport/physiology , Retroviridae/growth & development
9.
Adv Exp Med Biol ; 962: 247-264, 2017.
Article in English | MEDLINE | ID: mdl-28299662

ABSTRACT

The observation that the Runx genes act as targets for transcriptional activation by retroviral insertion identified a new family of dominant oncogenes. However, it is now clear that Runx genes are 'conditional' oncogenes whose over-expression is growth inhibitory unless accompanied by another event such as concomitant over-expression of MYC or loss of p53 function. Remarkably, while the oncogenic activities of either MYC or RUNX over-expression are suppressed while p53 is intact, the combination of both neutralises p53 tumour suppression in vivo by as yet unknown mechanisms. Moreover, there is emerging evidence that endogenous, basal RUNX activity is important to maintain the viability and proliferation of MYC-driven lymphoma cells. There is also growing evidence that the human RUNX genes play a similar conditional oncogenic role and are selected for over-expression in end-stage cancers of multiple types. Paradoxically, reduced RUNX activity can also predispose to cell immortalisation and transformation, particularly by mutant Ras. These apparently conflicting observations may be reconciled in a stage-specific model of RUNX involvement in cancer. A question that has yet to be fully addressed is the extent to which the three Runx genes are functionally redundant in cancer promotion and suppression.


Subject(s)
Core Binding Factor alpha Subunits/genetics , Neoplasms/genetics , Oncogenes/genetics , Retroviridae/growth & development , Animals , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Humans , Mice , Tumor Suppressor Protein p53/genetics
10.
Cell Host Microbe ; 20(6): 702-704, 2016 Dec 14.
Article in English | MEDLINE | ID: mdl-27978432

ABSTRACT

Integration is a key feature of the retroviral life cycle. This process involves packaging of the viral genome into chromatin, which is often assumed to occur as a post-integration step. In this issue of Cell Host & Microbe, Wang and colleagues (Wang et al., 2016) show that chromatinization occurs before integration, raising new questions about the role of histones in retroviral integration and transcription.


Subject(s)
Retroviridae/growth & development , Retroviridae/genetics , Virus Assembly , Virus Integration/genetics , Acetylation , Animals , Capsid Proteins/metabolism , Cell Line, Tumor , Chromatin/genetics , Chromatin/virology , DNA, Viral/genetics , DNA, Viral/physiology , Embryonal Carcinoma Stem Cells/virology , Epigenomics , Fibroblasts , Gene Expression Regulation, Viral , Histones/metabolism , Histones/physiology , Humans , Infections/metabolism , Life Cycle Stages , Mice , Mouse Embryonic Stem Cells/virology , Nucleocapsid Proteins/metabolism , Retroviridae Infections/therapy , Retroviridae Infections/virology , Transcription, Genetic , Virus Integration/physiology
11.
mBio ; 7(1): e02025-15, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26861021

ABSTRACT

A fascinating aspect of retroviruses is their tendency to nonrandomly incorporate host cell RNAs into virions. In addition to the specific tRNAs that prime reverse transcription, all examined retroviruses selectively package multiple host cell noncoding RNAs (ncRNAs). Many of these ncRNAs appear to be encapsidated shortly after synthesis, before assembling with their normal protein partners. Remarkably, although some packaged ncRNAs, such as pre-tRNAs and the spliceosomal U6 small nuclear RNA (snRNA), were believed to reside exclusively within mammalian nuclei, it was demonstrated recently that the model retrovirus murine leukemia virus (MLV) packages these ncRNAs from a novel pathway in which unneeded nascent ncRNAs are exported to the cytoplasm for degradation. The finding that retroviruses package forms of ncRNAs that are rare in cells suggests several hypotheses for how these RNAs could assist retrovirus assembly and infectivity. Moreover, recent experiments in several laboratories have identified additional ways in which cellular ncRNAs may contribute to the retrovirus life cycle. This review focuses on the ncRNAs that are packaged by retroviruses and the ways in which both encapsidated ncRNAs and other cellular ncRNAs may contribute to retrovirus replication.


Subject(s)
RNA, Nuclear/metabolism , RNA, Untranslated/metabolism , Retroviridae/physiology , Virus Replication , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Mice , Retroviridae/genetics , Retroviridae/growth & development , Virion/genetics , Virion/physiology , Virus Assembly/genetics
12.
Cold Spring Harb Protoc ; 2016(2): pdb.prot078337, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26832680

ABSTRACT

Since the introduction of retroviral vector technology, permanent genetic marking of cells has considerably contributed to the understanding of different physiological and disease processes in vivo. Recent marking strategies aim to elucidate the contribution of cells on the clonal level, and the advent of fluorescent proteins has opened new avenues for the in vivo analysis of gene-marked cells. Gene-modified cells are easily identifiable (e.g., via the introduced fluorescent protein) within whole organ structures, allowing one to measure the contribution of transduced cells to malignant outgrowth. In our laboratory, we use the tetracycline-inducible system to study oncogene cooperation in metastatic progression. We use bicistronic retroviruses expressing the tetracycline transactivator (tTA) and the candidate gene (MIT-gene) or the tTA alone (MIT-Rx) to infect primary mammary cells from mice harboring tetracycline-inducible transgenes. This allows for constitutive expression of the candidate gene and tTA-dependent expression of the inducible oncogene. We also use MIG-based vectors, which allow for constitutive expression of the candidate gene and a green fluorescent protein. Here we describe how to produce retroviral particles carrying both MIT- and MIG-based vectors. Because of the fragility of the retroviral envelope, we do not attempt to concentrate the virus, and we directly use packaging cell media to infect primary epithelial cells (either normal or tumor). Infected cells can be transplanted into recipient mice to investigate metastatic colonization.


Subject(s)
Genetic Vectors , Retroviridae/growth & development , Stem Cells/physiology , Virus Cultivation , Animals , Cells, Cultured , Gene Expression Regulation , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Retroviridae/genetics , Transduction, Genetic
13.
J Virol ; 90(7): 3400-10, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26764007

ABSTRACT

UNLABELLED: TRIM5α is an interferon-inducible retroviral restriction factor that prevents infection by inducing the abortive disassembly of capsid cores recognized by its C-terminal PRY/SPRY domain. The mechanism by which TRIM5α mediates the disassembly of viral cores is poorly understood. Previous studies demonstrated that proteasome inhibitors abrogate the ability of TRIM5α to induce premature core disassembly and prevent reverse transcription; however, viral infection is still inhibited, indicating that the proteasome is partially involved in the restriction process. Alternatively, we and others have observed that TRIM5α associates with proteins involved in autophagic degradation pathways, and one recent study found that autophagic degradation is required for the restriction of retroviruses by TRIM5α. Here, we show that TRIM5α is basally degraded via autophagy in the absence of restriction-sensitive virus. We observe that the autophagy markers LC3b and lysosome-associated membrane protein 2A (LAMP2A) localize to a subset of TRIM5α cytoplasmic bodies, and inhibition of lysosomal degradation with bafilomycin A1 increases this association. To test the requirement for macroautophagy in restriction, we examined the ability of TRIM5α to restrict retroviral infection in cells depleted of the autophagic mediators ATG5, Beclin1, and p62. In all cases, restriction of retroviruses by human TRIM5α, rhesus macaque TRIM5α, and owl monkey TRIM-Cyp remained potent in cells depleted of these autophagic effectors by small interfering RNA (siRNA) knockdown or clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 genome editing. Collectively, these results are consistent with observations that the turnover of TRIM5α proteins is sensitive to autophagy inhibition; however, the data presented here do not support observations that the inhibition of autophagy abrogates retroviral restriction by TRIM5 proteins. IMPORTANCE: Restriction factors are a class of proteins that inhibit viral replication. Following fusion of a retrovirus with a host cell membrane, the retroviral capsid is released into the cytoplasm of the target cell. TRIM5α inhibits retroviral infection by promoting the abortive disassembly of incoming retroviral capsid cores; as a result, the retroviral genome is unable to traffic to the nucleus, and the viral life cycle is extinguished. In the process of restriction, TRIM5α itself is degraded by the proteasome. However, in the present study, we have shown that in the absence of a restriction-sensitive virus, TRIM5α is degraded by both proteasomal and autophagic degradation pathways. Notably, we observed that restriction of retroviruses by TRIM5α does not require autophagic machinery. These data indicate that the effector functions of TRIM5α can be separated from its degradation and may have further implications for understanding the mechanisms of other TRIM family members.


Subject(s)
Autophagy/genetics , Carrier Proteins/metabolism , Retroviridae Infections/virology , Retroviridae/growth & development , Viral Core Proteins/metabolism , Virus Replication/genetics , Animals , Antiviral Restriction Factors , Aotidae , Apoptosis Regulatory Proteins/genetics , Autophagy-Related Protein 5 , Beclin-1 , Carrier Proteins/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , HeLa Cells , Humans , Lysosomal Membrane Proteins/metabolism , Macaca mulatta , Macrolides/pharmacology , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Proteolysis , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Retroviridae/genetics , Retroviridae Infections/immunology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
14.
Biotechnol Bioeng ; 113(1): 150-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26134455

ABSTRACT

Many mammalian cell lines used in the manufacturing of biopharmaceuticals exhibit high glycolytic flux predominantly channeled to the production of lactate. The accumulation of lactate in culture reduces cell viability and may also decrease product quality. In this work, we engineered a HEK 293 derived cell line producing a recombinant gene therapy retroviral vector, by down-regulating hypoxia inducible factor 1 (HIF1) and pyruvate dehydrogenase kinase (PDK). Specific productivity of infectious viral titers could be increased more than 20-fold for single gene knock-down (HIF1 or PDK) and more than 30-fold under combined down-regulation. Lactate production was reduced up to 4-fold. However, the reduction in lactate production, alone, was not sufficient to enhance the titer: high-titer clones also showed significant enrollment of metabolic routes not related to lactate production. Transcriptome analysis indicated activation of biological amines metabolism, detoxification routes, including glutathione metabolism, pentose phosphate pathway, glycogen biosynthesis and amino acid catabolism. The latter were validated by enzyme activity assays and metabolite profiling, respectively. High-titer clones also presented substantially increased transcript levels of the viral genes expression cassettes. The results herein presented demonstrate the impact of HIF1 and PDK down-regulation on the production performance of a mammalian cell line, reporting one of the highest fold-increase in specific productivity of infectious virus titers achieved by metabolic engineering. They additionally highlight the contribution of secondary pathways, beyond those related to lactate production, that can be also explored to pursue improved metabolic status favoring a high-producing phenotype.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Lactic Acid/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Retroviridae/growth & development , Viral Load , Virus Cultivation/methods , Cell Line , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
15.
Cold Spring Harb Protoc ; 2015(7): 679-84, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26134903

ABSTRACT

Stem cell transplantation is well established in humans for the treatment of hematopoietic disease, including hematopoietic malignancies. Similar direct transplant procedures can readily be performed in mice; these procedures can be paired with retroviral infection to introduce exogenous genes or to silence endogenous genes in a subset of cells in the murine hematopoietic system. The resulting mice are chimeric for cells bearing a specific alteration. This approach has the advantage of examining tumorigenesis on a largely wild-type background (if only a subset of cells are infected), a situation that more accurately parallels the human situation. Additionally, tumor development occurs within the appropriate native microenvironment. Here, we describe the isolation and retroviral infection of hematopoietic stem cells (HSCs), as well as the reconstitution and monitoring of tumor formation in lethally irradiated recipient mice. This protocol requires a source of long-term HSCs; these can include either stimulated adult bone marrow or fetal liver­the site of primitive hematopoiesis.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/virology , Liver/physiology , Organisms, Genetically Modified , Retroviridae/genetics , Stem Cells/physiology , Transduction, Genetic , Animals , Disease Models, Animal , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Mice , Retroviridae/growth & development
16.
Cold Spring Harb Protoc ; 2015(7): 685-8, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26134904

ABSTRACT

Hepatic progenitor cells isolated from fetal liver can be transplanted into recipient mice to reconstitute an organ system. Retroviral infection can be used to introduce putative oncogenes or short-hairpin RNAs (shRNAs) targeting putative tumor suppressors into the cells; reconstituted mice then develop livers that are chimeric for cells bearing a specific alteration. This approach has the advantage of examining tumorigenesis on a largely wild-type background (if only a subset of cells are infected), a situation that more accurately parallels the human situation. Additionally, tumor development occurs within the appropriate native microenvironment. Here, we describe the isolation of hepatic progenitor cells, as well as the reconstitution and tumor monitoring of recipient mice.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/virology , Liver/physiology , Organisms, Genetically Modified , Retroviridae/genetics , Stem Cells/physiology , Transduction, Genetic , Animals , Disease Models, Animal , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Mice , Retroviridae/growth & development
17.
Virology ; 479-480: 297-309, 2015 May.
Article in English | MEDLINE | ID: mdl-25680625

ABSTRACT

Many important aspects of human retroviral infections cannot be fully evaluated using only in vitro systems or unmodified animal models. An alternative approach involves the use of humanized mice, which consist of immunodeficient mice that have been transplanted with human cells and/or tissues. Certain humanized mouse models can support robust infection with human retroviruses including different strains of human immunodeficiency virus (HIV) and human T cell leukemia virus (HTLV). These models have provided wide-ranging insights into retroviral biology, including detailed information on primary infection, in vivo replication and pathogenesis, latent/persistent reservoir formation, and novel therapeutic interventions. Here we describe the humanized mouse models that are most commonly utilized to study retroviral infections, and outline some of the important discoveries that these models have produced during several decades of intensive research.


Subject(s)
Disease Models, Animal , Host-Pathogen Interactions , Retroviridae Infections/pathology , Retroviridae Infections/virology , Retroviridae/physiology , Animals , Humans , Mice, SCID , Retroviridae/growth & development , Retroviridae/pathogenicity , Virus Latency , Virus Replication
18.
Hum Gene Ther Clin Dev ; 25(3): 178-85, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25073060

ABSTRACT

Human immunodeficiency virus type 1-derived lentiviral vectors (LVs) are becoming major tools for gene transfer approaches. Several gene therapy clinical studies involving LVs are currently ongoing. Industrial production of clinical-grade LVs is therefore an important challenge. Some improvements in LV production protocols have already been possible by acting on multiple steps of the production process like transfection, cell culture, or media optimizations. Yet, the effects of physicochemical parameters such as pH remain poorly studied. Mammalian cell cultures are generally performed at neutral pH, which may not be the optimal condition to produce high quantities of LVs with optimal infectious properties. In this study, we showed that lentiviral transient production in HEK293T cells is inversely dependent on the pH value of the harvesting medium. Infectious and physical titers of LVs pseudotyped with GALVTR or VSV-G glycoproteins are enhanced by two- to threefold at pH 6 compared with neutral conditions. pH 6-produced LVs are highly infectious on cell lines but also on relevant primary target cells like hCD34+ hematopoietic stem/progenitor cells. GALVTR-LV particles produced at pH 6 are highly stable at 37 °C and resistant to multiple freeze-thaw cycles. Higher levels of expression of intracellular pr55gag polyproteins are observed within HEK293T producer cells cultured at pH 6. The positive effect of pH 6 conditions is also observed for moloney-derived retroviral vectors produced from NIH-3T3 fibroblasts, arguing that the mildly acidic pH effect is not limited to the lentivirus genus and is reproducible in various producer cell lines. This observation may help us in the design of more effective LV production protocols for clinical applications.


Subject(s)
Genetic Vectors/biosynthesis , Lentivirus/growth & development , Retroviridae/growth & development , Antigens, CD34/metabolism , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , HEK293 Cells , Hematopoietic Stem Cells , Humans , Hydrogen-Ion Concentration , Transduction, Genetic , Transfection , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
19.
Biotechnol Bioeng ; 109(5): 1269-79, 2012 May.
Article in English | MEDLINE | ID: mdl-22179842

ABSTRACT

The manufacture of enveloped virus, particularly retroviral (RV) and lentiviral (LV) vectors, faces the challenge of low titers that are aggravated under serum deprivation culture conditions. Also, the scarce knowledge on the biochemical pathways related with virus production is still limiting the design of rational strategies for improved production yields. This work describes the adaptation to serum deprivation of two human RV packaging cell lines, 293 FLEX and Te Fly and its effects on lipid biosynthetic pathways and infectious vector production. Total lipid content as well as cellular cholesterol were quantified and lipid biosynthesis was assessed by (13)C-NMR spectroscopy; changes in gene expression of lipid biosynthetic enzymes were also evaluated. The effects of adaptation to serum deprivation in lipid biosynthesis were cell line specific and directly correlated with infectious virus titers: 293 FLEX cells faced severe lipid starvation-up to 50% reduction in total lipid content-along with a 68-fold reduction in infectious vector titers; contrarily, Te Fly cells were able to maintain identical levels of total lipid content by rising de novo lipid biosynthesis, particularly for cholesterol-50-fold increase-with the consequent recovery of infectious vector productivities. Gene expression analysis of lipid biosynthetic enzymes further confirmed cholesterol production pathway to be prominently up-regulated under serum deprivation conditions for Te Fly cells, providing a genotype-phenotype validation for enhanced cholesterol synthesis. These results highlight lipid metabolism dynamics and the ability to activate lipid biosynthesis under serum deprivation as an important feature for high retroviral titers. Mechanisms underlying virus production and its relationship with lipid biosynthesis, with special focus on cholesterol, are discussed as potential targets for cellular metabolic engineering.


Subject(s)
Cell Proliferation , Culture Media, Serum-Free/chemistry , Genetic Vectors , Lipid Metabolism , Retroviridae/growth & development , Biosynthetic Pathways/genetics , Cell Line , Cytosol/chemistry , Gene Expression Profiling , Humans , Magnetic Resonance Spectroscopy , Viral Load
20.
Biotechnol Bioeng ; 108(11): 2623-33, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21656710

ABSTRACT

Retroviral-derived biopharmaceuticals (RV) target numerous therapeutic applications, from gene therapy to virus-like particle (rVLP)-based vaccines. During particle formation, beside the pseudotyped envelope proteins, RV can incorporate proteins derived from the virus producer cells (VPC). This may be detrimental by reducing the amounts of the pseudotyped envelope and/or by incorporating protein capable of inducing immune responses when non-human VPC are used. Manipulating the repertoire of VPC proteins integrated onto the vector structure is an underexplored territory and should provide valuable insights on potential targets to improve vector pharmacokinetic and pharmacodynamic properties. In this work, human HEK 293 cells producing retrovirus-like particles (rVLPs) and infectious RV vectors were used to prove the concept of customizing RV composition by manipulating cellular protein content. The tetraspanin CD81 was chosen since it is significantly incorporated in the RV membrane, conferring to the vector significant immunogenicity when used in mice. RNA interference-mediated by shRNA lentiviral vector transduction was efficiently used to silence CD81 expression (up to 99%) and the rVLPs produced by knocked-down cells lack CD81. Silenced clones were analyzed for cell proliferation, morphological changes, susceptibility to oxidative stress conditions, and rVLP productivities. The results showed that the down-regulation of VPC proteins requires close monitoring for possible side effects on cellular production performance. Yet, they confirm that it is possible to change the composition of host-derived immunogens in RV by altering cellular protein content with no detriment for vector productivity and titers. This constitutes an important manipulation tool in vaccinology--by exploiting the potential adjuvant effect of VPC proteins or using them as fusion agents to other proteins of interest to be exposed on the vector membrane--and in gene therapy, by reducing the immunogenicity of RV-based vector and enhancing in vivo half-life. Such tools can also be applied to lentiviral or other enveloped viral vectors.


Subject(s)
Biological Products/chemistry , Down-Regulation , Genetic Vectors , Retroviridae/chemistry , Retroviridae/genetics , Tetraspanin 28/analysis , Animals , Biological Products/administration & dosage , Biological Products/isolation & purification , Cell Line , Gene Knockdown Techniques/methods , Gene Silencing , Humans , Mice , Retroviridae/growth & development , Retroviridae/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...