Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(5): 1311-1327, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38449314

ABSTRACT

While studying transgene expression after systemic administration of lentiviral vectors, we found that splenic B cells are robustly transduced, regardless of the types of pseudotyped envelope proteins. However, the administration of two different pseudotypes resulted in transduction of two distinct B cell populations, suggesting that each pseudotype uses unique and specific receptors for its attachment and entry into splenic B cells. Single-cell RNA sequencing analysis of the transduced cells demonstrated that different pseudotypes transduce distinct B cell subpopulations characterized by specific B cell receptor (BCR) genotypes. Functional analysis of the BCRs of the transduced cells demonstrated that BCRs specific to the pseudotyping envelope proteins mediate viral entry, enabling the vectors to selectively transduce the B cell populations that are capable of producing antibodies specific to their envelope proteins. Lentiviral vector entry via the BCR activated the transduced B cells and induced proliferation and differentiation into mature effectors, such as memory B and plasma cells. BCR-mediated viral entry into clonally specific B cell subpopulations raises new concepts for understanding the biodistribution of transgene expression after systemic administration of lentiviral vectors and offers new opportunities for BCR-targeted gene delivery by pseudotyped lentiviral vectors.


Subject(s)
B-Lymphocytes , Genetic Vectors , Lentivirus , Receptors, Antigen, B-Cell , Transduction, Genetic , Transgenes , Viral Envelope Proteins , Lentivirus/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Animals , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Tropism , Humans , Virus Internalization
2.
PLoS Biol ; 20(11): e3001851, 2022 11.
Article in English | MEDLINE | ID: mdl-36346780

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), responsible for the Coronavirus Disease 2019 (COVID-19) pandemic, causes respiratory failure and damage to multiple organ systems. The emergence of viral variants poses a risk of vaccine failures and prolongation of the pandemic. However, our understanding of the molecular basis of SARS-CoV-2 infection and subsequent COVID-19 pathophysiology is limited. In this study, we have uncovered a critical role for the evolutionarily conserved Hippo signaling pathway in COVID-19 pathogenesis. Given the complexity of COVID-19-associated cell injury and immunopathogenesis processes, we investigated Hippo pathway dynamics in SARS-CoV-2 infection by utilizing COVID-19 lung samples and human cell models based on pluripotent stem cell-derived cardiomyocytes (PSC-CMs) and human primary lung air-liquid interface (ALI) cultures. SARS-CoV-2 infection caused activation of the Hippo signaling pathway in COVID-19 lung and in vitro cultures. Both parental and Delta variant of concern (VOC) strains induced Hippo pathway. The chemical inhibition and gene knockdown of upstream kinases MST1/2 and LATS1 resulted in significantly enhanced SARS-CoV-2 replication, indicating antiviral roles. Verteporfin, a pharmacological inhibitor of the Hippo pathway downstream transactivator, YAP, significantly reduced virus replication. These results delineate a direct antiviral role for Hippo signaling in SARS-CoV-2 infection and the potential for this pathway to be pharmacologically targeted to treat COVID-19.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Hippo Signaling Pathway , Antiviral Agents/pharmacology
3.
Microbiol Spectr ; 10(5): e0113722, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36169338

ABSTRACT

Zika virus (ZIKV), a mosquito-borne human pathogen, causes dire congenital brain developmental abnormalities in children of infected mothers. The global health crisis precipitated by this virus has led to a concerted effort to develop effective therapies and prophylactic measures although, unfortunately, not very successfully. The error-prone nature of RNA viral genome replication tends to promote evolution of novel viral strains, which could cause epidemics and pandemics. As such, our objective was to develop a safe and effective replication-deficient ZIKV vector-based vaccine candidate. We approached this by generating a ZIKV vector containing only the nonstructural (NS) 5'-untranslated (UTR)-NS-3' UTR sequences, with the structural proteins capsid (C), precursor membrane (prM), and envelope (E) (CprME) used as a packaging system. We efficiently packaged replication-deficient Zika vaccine particles in human producer cells and verified antigen expression in vitro. In vivo studies showed that, after inoculation in neonatal mice, the Zika vaccine candidate (ZVAX) was safe and did not produce any replication-competent revertant viruses. Immunization of adult, nonpregnant mice showed that ZVAX protected mice from lethal challenge by limiting viral replication. We then evaluated the safety and efficacy of ZVAX in pregnant mice, where it was shown to provide efficient maternal and fetal protection against Zika disease. Mass cytometry analysis showed that vaccinated pregnant animals had high levels of splenic CD8+ T cells and effector memory T cell responses with reduced proinflammatory cell responses, suggesting that endogenous expression of NS proteins by ZVAX induced cellular immunity against ZIKV NS proteins. We also investigated humoral immunity against ZIKV, which is potentially induced by viral proteins present in ZVAX virions. We found no significant difference in neutralizing antibody titer in vaccinated or unvaccinated challenged animals; therefore, it is likely that cellular immunity plays a major role in ZVAX-mediated protection against ZIKV infection. In conclusion, we demonstrated ZVAX as an effective inducer of protective immunity against ZIKV, which can be further evaluated for potential prophylactic application in humans. IMPORTANCE This research is important as it strives to address the critical need for effective prophylactic measures against the outbreak of Zika virus (ZIKV) and outlines an important vaccine technology that could potentially be used to induce immune responses against other pandemic-potential viruses.


Subject(s)
Viral Vaccines , Zika Virus Infection , Zika Virus , Pregnancy , Female , Child , Mice , Humans , Animals , Zika Virus/genetics , Zika Virus Infection/prevention & control , CD8-Positive T-Lymphocytes , 3' Untranslated Regions , Viral Vaccines/genetics , Antibodies, Viral , Viral Envelope Proteins/genetics , Mosquito Vectors , Antibodies, Neutralizing , Disease Models, Animal
4.
bioRxiv ; 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35441167

ABSTRACT

SARS-CoV-2, responsible for the COVID-19 pandemic, causes respiratory failure and damage to multiple organ systems. The emergence of viral variants poses a risk of vaccine failures and prolongation of the pandemic. However, our understanding of the molecular basis of SARS-CoV-2 infection and subsequent COVID-19 pathophysiology is limited. In this study, we have uncovered a critical role for the evolutionarily conserved Hippo signaling pathway in COVID-19 pathogenesis. Given the complexity of COVID-19 associated cell injury and immunopathogenesis processes, we investigated Hippo pathway dynamics in SARS-CoV-2 infection by utilizing COVID-19 lung samples, and human cell models based on pluripotent stem cell-derived cardiomyocytes (PSC-CMs) and human primary lung air-liquid interface (ALI) cultures. SARS-CoV-2 infection caused activation of the Hippo signaling pathway in COVID-19 lung and in vitro cultures. Both parental and Delta variant of concern (VOC) strains induced Hippo pathway. The chemical inhibition and gene knockdown of upstream kinases MST1/2 and LATS1 resulted in significantly enhanced SARS-CoV-2 replication, indicating antiviral roles. Verteporfin a pharmacological inhibitor of the Hippo pathway downstream transactivator, YAP, significantly reduced virus replication. These results delineate a direct antiviral role for Hippo signaling in SARS-CoV-2 infection and the potential for this pathway to be pharmacologically targeted to treat COVID-19.

5.
Viruses ; 13(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34834920

ABSTRACT

Zika virus (ZIKV) is a re-emerging flavivirus that has caused large-scale epidemics. Infection during pregnancy can lead to neurologic developmental abnormalities in children. There is no approved vaccine or therapy for ZIKV. To uncover cellular pathways required for ZIKV that can be therapeutically targeted, we transcriptionally upregulated all known human coding genes with an engineered CRISPR-Cas9 activation complex in human fibroblasts deficient in interferon (IFN) signaling. We identified Ras homolog family member V (RhoV) and WW domain-containing transcription regulator 1 (WWTR1) as proviral factors, and found them to play important roles during early ZIKV infection in A549 cells. We then focused on RhoV, a Rho GTPase with atypical terminal sequences and membrane association, and validated its proviral effects on ZIKV infection and virion production in SNB-19 cells. We found that RhoV promotes infection of some flaviviruses and acts at the step of viral entry. Furthermore, RhoV proviral effects depend on the complete GTPase cycle. By depleting Rho GTPases and related proteins, we identified RhoB and Pak1 as additional proviral factors. Taken together, these results highlight the positive role of RhoV in ZIKV infection and confirm CRISPR activation as a relevant method to identify novel host-pathogen interactions.


Subject(s)
GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Zika Virus Infection/enzymology , Zika Virus/physiology , rhoB GTP-Binding Protein/metabolism , A549 Cells , CRISPR-Cas Systems , GTP-Binding Proteins/genetics , Humans , Neoplasm Proteins/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Virus Internalization , Virus Replication , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/virology , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rhoB GTP-Binding Protein/genetics
6.
Virology ; 560: 17-33, 2021 08.
Article in English | MEDLINE | ID: mdl-34020328

ABSTRACT

Envelope phosphatidylserine (PtdSer) and phosphatidylethanolamine (PtdEtr) have been shown to mediate binding of enveloped viruses. However, commonly used PtdSer binding molecules such as Annexin V cannot block PtdSer-mediated viral infection. Lack of reagents that can conceal envelope PtdSer and PtdEtr and subsequently inhibit infection hinders elucidation of the roles of the envelope phospholipids in viral infection. Here, we developed sTIM1dMLDR801, a reagent capable of blocking PtdSer- and PtdEtr-dependent infection of enveloped viruses. Using sTIM1dMLDR801, we found that envelope PtdSer and/or PtdEtr can support ZIKV infection of not only human but also mosquito cells. In a mouse model for ZIKV infection, sTIM1dMLDR801 reduced ZIKV load in serum and the spleen, indicating envelope PtdSer and/or PtdEtr support in viral infection in vivo. sTIM1dMLDR801 will enable elucidation of the roles of envelope PtdSer and PtdEtr in infection of various virus species, thereby facilitating identification of their receptors and transmission mechanisms.


Subject(s)
Antiviral Agents/pharmacology , Phosphatidylethanolamines/antagonists & inhibitors , Phosphatidylserines/antagonists & inhibitors , Virus Attachment/drug effects , Virus Internalization/drug effects , Zika Virus/drug effects , A549 Cells , Animals , Cell Line , Chlorocebus aethiops , Culicidae/virology , Female , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Interferon alpha-beta/genetics , Vero Cells , Viral Envelope/metabolism , Viral Load/drug effects , Zika Virus/growth & development , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology , Zika Virus Infection/transmission , Axl Receptor Tyrosine Kinase
7.
PeerJ ; 9: e10725, 2021.
Article in English | MEDLINE | ID: mdl-33552733

ABSTRACT

The risk for breast cancer is significantly reduced in persons who engage in greater amounts of physical activity, and greater physical activity before or after diagnosis associates with reduced disease-specific mortality. Previous mechanistic studies indicate that components of innate immunity can mediate an inhibitory effect of physical activity on several types of tumor. However, in breast cancer specifically, the myeloid compartment of innate immunity is thought to exhibit high propensity for an immunosuppressive role that obstructs anti-tumor immunity. Thus, we tested the notion that greater physical activity alters mononuclear phagocytes in mammary tissue when inhibiting nascent tumor in a murine model of breast cancer. To model greater physical activity, we placed an angled running wheel in each mouse's home cage for two weeks before tumor engraftment with EO771 mammary cancer cells that express luciferase for bioluminescent detection. Fully immunocompetent mice and mice with compromised adaptive immunity showed significantly less mammary tumor signal when given access to running wheels, although the effect size was smaller in this latter group. To investigate the role of the myeloid compartment, mononuclear phagocytes were ablated by systemic injection of clodronate liposomes at 24 h before tumor engraftment and again at the time of tumor engraftment, and this treatment reversed the inhibition in wheel running mice. However, clodronate also inhibited mammary tumor signal in sedentary mice, in conjunction with an expected decrease in gene and protein expression of the myeloid antigen, F4/80 (Adgre1), in mammary tissue. Whole transcriptome digital cytometry with CIBERSORTx was used to analyze myeloid cell populations in mammary tissue following voluntary wheel running and clodronate treatment, and this approach found significant changes in macrophage and monocyte populations. In exploratory analyses, whole transcriptome composite scores for monocytic myeloid-derived suppressor cell (M-MDSC), macrophage lactate timer, and inflammation resolution gene expression programs were significantly altered. Altogether, the results support the hypothesis that physical activity inhibits nascent mammary tumor growth by enhancing the anti-tumor potential of mononuclear phagocytes in mammary tissue.

8.
Cell Rep Med ; 1(4): 100052, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32835305

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is defined by respiratory symptoms, but cardiac complications including viral myocarditis are also prevalent. Although ischemic and inflammatory responses caused by COVID-19 can detrimentally affect cardiac function, the direct impact of SARS-CoV-2 infection on human cardiomyocytes is not well understood. Here, we utilize human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as a model to examine the mechanisms of cardiomyocyte-specific infection by SARS-CoV-2. Microscopy and RNA sequencing demonstrate that SARS-CoV-2 can enter hiPSC-CMs via ACE2. Viral replication and cytopathic effect induce hiPSC-CM apoptosis and cessation of beating after 72 h of infection. SARS-CoV-2 infection activates innate immune response and antiviral clearance gene pathways, while inhibiting metabolic pathways and suppressing ACE2 expression. These studies show that SARS-CoV-2 can infect hiPSC-CMs in vitro, establishing a model for elucidating infection mechanisms and potentially a cardiac-specific antiviral drug screening platform.

9.
Am J Pathol ; 190(4): 844-861, 2020 04.
Article in English | MEDLINE | ID: mdl-32035058

ABSTRACT

Zika virus (ZIKV) is a reemerging human pathogen that causes congenital abnormalities, including microcephaly and eye disease. The cellular/molecular basis of ZIKV and host interactions inducing ocular and neuronal pathogenesis are unclear. Herein, we noted that the Hippo/Salvador-Warts-Hippo signaling pathway, which controls organ size through progenitor cell proliferation and differentiation, is dysregulated after ZIKV infection. In human fetal retinal pigment epithelial cells, there is an early induction of transcriptional coactivator, Yes-associated protein (YAP), which is later degraded with a corresponding activation of the TANK binding kinase 1/interferon regulatory factor 3 type I interferon pathway. YAP/transcriptional co-activator with a PDZ-binding domain (TAZ) silencing results in reduced ZIKV replication, indicating a direct role of Hippo pathway in regulating ZIKV infection. Using an in vivo Ifnar1-/- knockout mouse model, ZIKV infection was found to reduce YAP/TAZ protein levels while increasing phosphorylated YAP Ser127 in the retina and brain. Hippo pathway is activated in major cellular components of the blood-brain barrier, including endothelial cells and astrocytes. In addition, this result suggests AMP-activated protein kinase signaling pathway's role in regulating YAP/TAZ in ZIKV-infected cells. These data demonstrate that ZIKV infection might initiate a cross talk among AMP-activated protein kinase-Hippo-TBK1 pathways, which could regulate antiviral and energy stress responses during oculoneuronal inflammation.


Subject(s)
Inflammation/pathology , Neurodegenerative Diseases/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Interferon alpha-beta/physiology , Virus Replication , Zika Virus Infection/complications , Zika Virus/isolation & purification , Animals , Hippo Signaling Pathway , Inflammation/virology , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/virology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Zika Virus Infection/virology
10.
Cell Commun Signal ; 17(1): 132, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31638994

ABSTRACT

Phosphatidylserine (PtdSer) is usually present only in the inner leaf of the lipid bilayers of the cell membrane, but is exposed on the outer leaf when cells are activated and/or die. Exposure of PtdSer has physiological functions. For example, the PtdSer exposed on dead cells can serve as "eat-me signals" for phagocytes to clear dead cells by phagocytosis, which prevents autoimmune reactions and inflammation. HIV-1 induces PtdSer exposure on infected and target cells and it also exposes PtdSer on its envelope. Recent studies showed that PtdSer exposed on the HIV-1 envelope and infected and target cells can facilitate or inhibit multiple steps of HIV-1 replication.At the virus binding and entry steps, interaction of the envelope PtdSer and the host's PtdSer-binding molecules can enhance HIV-1 infection of cells by facilitating virus attachment. At the virus budding step, HIV-1 can be trapped on the cell surface by one family of PtdSer-binding receptors, T-cell immunoglobulin mucin domain proteins (TIM)-1, 3, and 4 expressed on virus producer cells. Although this trapping can inhibit release of HIV-1, one of the HIV-1 accessory gene products, Negative Factor (Nef), can counteract virus trapping by TIM family receptors (TIMs) by inducing the internalization of these receptors. HIV-1 infection can induce exposure of PtdSer on infected cells by inducing cell death. A soluble PtdSer-binding protein in serum, protein S, bridges PtdSer exposed on HIV-1-infected cells and a receptor tyrosine kinase, Mer, expressed on macrophages and mediate phagocytic clearance of HIV-1 infected cells. HIV-1 can also induce exposure of PtdSer on target cells at the virus binding step. Binding of HIV-1 envelope proteins to its receptor (CD4) and co-receptors (CXCR4 or CCR5) elicit signals that induce PtdSer exposure on target cells by activating TMEM16F, a phospholipid scramblase. PtdSer exposed on target cells enhances HIV-1 infection by facilitating fusion between the viral envelope and target cell membrane. Because various other phospholipid channels mediating PtdSer exposure have recently been identified, it will be of interest to examine how HIV-1 actively interacts with these molecules to manipulate PtdSer exposure levels on cells and viral envelope to support its replication.


Subject(s)
Cell Membrane/metabolism , HIV-1/physiology , Phosphatidylserines/metabolism , Virus Replication , Animals , Humans , Macrophages/cytology , Macrophages/virology , Virus Internalization
11.
Virology ; 525: 170-181, 2018 12.
Article in English | MEDLINE | ID: mdl-30290312

ABSTRACT

Conjugating certain types of lentiviral vectors with targeting ligands can redirect the vectors to specifically transduce desired cell types. However, extensive genetic and/or biochemical manipulations are required for conjugation, which hinders applications for targeting lentiviral vectors for broader research fields. We developed envelope proteins fused with biotin-binding molecules to conjugate the pseudotyped vectors with biotinylated targeting molecules by simply mixing them. The envelope proteins fused with the monomeric, but not tetrameric, biotin-binding molecules can pseudotype lentiviral vectors and be conjugated with biotinylated targeting ligands. The conjugation is stable enough to redirect lentiviral transduction in the presence of serum, indicating their potential in in vivo . When a signaling molecule is conjugated with the vector, the conjugation facilitates transduction and signaling in a receptor-specific manner. This simple method of ligand conjugation and ease of obtaining various types of biotinylated ligands will make targeted lentiviral transduction easily applicable to broad fields of research.


Subject(s)
Lentivirus/physiology , Biotin/metabolism , Biotinylation , Gene Expression Regulation, Viral , Gene Transfer Techniques , Genetic Vectors , Transduction, Genetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
12.
Virology ; 515: 176-190, 2018 02.
Article in English | MEDLINE | ID: mdl-29304470

ABSTRACT

Efferocytosis, the phagocytic clearance of apoptotic cells, can provide host protection against certain types of viruses by mediating phagocytic clearance of infected cells undergoing apoptosis. It is known that HIV-1 induces apoptosis and HIV-1-infected cells are efferocytosed by macrophages, although its molecular mechanisms are unknown. To elucidate the roles that efferocytosis of HIV-1-infected cells play in clearance of infected cells, we sought to identify molecules that mediate these processes. We found that protein S, present in human serum, and its homologue, Gas6, can mediate phagocytosis of HIV-1-infected cells by bridging receptor tyrosine kinase Mer, expressed on macrophages, to phosphatidylserine exposed on infected cells. Efferocytosis of live infected cells was less efficient than dead infected cells; however, a significant fraction of live infected cells were phagocytosed over 12h. Our results suggest that efferocytosis not only removes dead cells, but may also contribute to macrophage removal of live virus producing cells.


Subject(s)
HIV Infections/metabolism , HIV Infections/physiopathology , HIV-1/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Protein S/metabolism , Animals , HIV Infections/genetics , HIV Infections/virology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/cytology , Macrophages/metabolism , Phagocytosis , Protein S/genetics
13.
J Virol ; 88(8): 4275-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478428

ABSTRACT

UNLABELLED: We recently demonstrated that a soluble protein, Gas6, can facilitate viral entry by bridging viral envelope phosphatidylserine to Axl, a receptor tyrosine kinase expressed on target cells. The interaction between phosphatidylserine, Gas6, and Axl was originally shown to be a molecular mechanism through which phagocytes recognize phosphatidylserine exposed on dead cells. Since our initial report, several groups have confirmed that Axl/Gas6, as well as other phosphatidylserine receptors, facilitate entry of dengue, West Nile, and Ebola viruses. Virus binding by viral envelope phosphatidylserine is now a viral entry mechanism generalized to many families of viruses. In addition to Axl/Gas6, various molecules are known to recognize phosphatidylserine; however, the effects of these molecules on virus binding and entry have not been comprehensively evaluated and compared. In this study, we examined most of the known human phosphatidylserine-recognizing molecules, including MFG-E8, TIM-1, -3, and -4, CD300a, BAI1, and stabilin-1 and -2, for their abilities to facilitate virus binding and infection. Using pseudotyped lentiviral vectors, we found that a soluble phosphatidylserine-binding protein, MFG-E8, enhances transduction. Cell surface receptors TIM-1 and -4 also enhance virus binding/transduction. The extent of enhancement by these molecules varies, depending on the type of pseudotyping envelope proteins. Mutated MFG-E8, which binds viral envelope phosphatidylserine without bridging virus to cells, but, surprisingly, not annexin V, which has been used to block phagocytosis of dead cells by concealing phosphatidylserine, efficiently blocks these phosphatidylserine-dependent viral entry mechanisms. These results provide insight into understanding the role of viral envelope phosphatidylserine in viral infection. IMPORTANCE: Envelope phosphatidylserine has previously been shown to be important for replication of various envelope viruses, but details of this mechanism(s) were unclear. We were the first to report that a bifunctional serum protein, Gas6, bridges envelope phosphatidylserine to a cell surface receptor, Axl. Recent studies demonstrated that many envelope viruses, including vaccinia, dengue, West Nile, and Ebola viruses, utilize Axl/Gas6 to facilitate their entry, suggesting that the phosphatidylserine-mediated viral entry mechanism can be shared by various enveloped viruses. In addition to Axl/Gas6, various molecules are known to recognize phosphatidylserine; however, the effects of these molecules on virus binding and entry have not been comprehensively evaluated and compared. In this study, we examined most human phosphatidylserine-recognizing molecules for their abilities to facilitate viral infection. The results provide insights into the role(s) of envelope phosphatidylserine in viral infection, which can be applicable to the development of novel antiviral reagents that block phosphatidylserine-mediated viral entry.


Subject(s)
Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Virus Physiological Phenomena , Cell Line , Humans , Phagocytosis , Phosphatidylserines/metabolism , Receptors, Cell Surface/genetics , Receptors, Virus/genetics , Virus Attachment , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/virology , Virus Internalization , Viruses/genetics
14.
J Gene Med ; 16(1-2): 11-27, 2014.
Article in English | MEDLINE | ID: mdl-24436117

ABSTRACT

BACKGROUND: We previously developed an antibody-avidin fusion protein (ch128.1Av) specific for the human transferrin receptor 1 (TfR1; CD71) to be used as a delivery vector for cancer therapy and showed that ch128.1Av delivers the biotinylated plant toxin saporin-6 into malignant B cells. However, as a result of widespread expression of TfR1, delivery of the toxin to normal cells is a concern. Therefore, we explored the potential of a dual targeted lentiviral-mediated gene therapy strategy to restrict gene expression to malignant B cells. Targeting occurs through the use of ch128.1Av or its parental antibody without avidin (ch128.1) and through transcriptional regulation using an immunoglobulin promoter. METHODS: Flow cytometry was used to detect the expression of enhanced green fluorescent protein (EGFP) in a panel of cell lines. Cell viability after specific delivery of the therapeutic gene FCU1, a chimeric enzyme consisting of cytosine deaminase genetically fused to uracil phosphoribosyltransferse that converts the 5-fluorocytosine (5-FC) prodrug into toxic metabolites, was monitored using the MTS or WST-1 viability assay. RESULTS: We found that EGFP was specifically expressed in a panel of human malignant B-cell lines, but not in human malignant T-cell lines. EGFP expression was observed in all cell lines when a ubiquitous promoter was used. Furthermore, we show the decrease of cell viability in malignant plasma cells in the presence of 5-FC and the FCU1 gene. CONCLUSIONS: The present study demonstrates that gene expression can be restricted to malignant B cells and suggests that this dual targeted gene therapy strategy may help to circumvent the potential side effects of certain TfR1-targeted protein delivery approaches.


Subject(s)
Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocytes/cytology , Lentivirus/genetics , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Recombinant Fusion Proteins/genetics , Antibodies/genetics , Antibodies/immunology , Antigens, CD/biosynthesis , B-Lymphocytes/metabolism , Cell Line, Tumor , Cytosine Deaminase/genetics , Flucytosine/metabolism , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Green Fluorescent Proteins , HEK293 Cells , Humans , Jurkat Cells , Neoplasms/genetics , Neoplasms/therapy , Pentosyltransferases/genetics , Prodrugs/adverse effects , Prodrugs/therapeutic use , Promoter Regions, Genetic , Receptors, Transferrin/biosynthesis , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transduction, Genetic , Vesicular stomatitis Indiana virus/genetics
15.
J Virol ; 87(4): 2094-108, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192877

ABSTRACT

Sophisticated retargeting systems for lentiviral vectors have been developed in recent years. Most seek to suppress the viral envelope's natural tropism while modifying the receptor-binding domain such that its tropism is determined by the specificity of the engineered ligand-binding motif. Here we took advantage of the natural tropism of Nipah virus (NiV), whose attachment envelope glycoprotein has picomolar affinity for ephrinB2, a molecule proposed as a molecular marker of "stemness" (present on embryonic, hematopoietic, and neural stem cells) as well as being implicated in tumorigenesis of specific cancers. NiV entry requires both the fusion (F) and attachment (G) glycoproteins. Truncation of the NiV-F cytoplasmic tail (T5F) alone, combined with full-length NiV-G, resulted in optimal titers of NiV-pseudotyped particles (NiVpp) (∼10(6) IU/ml), even without ultracentrifugation. To further enhance the infectivity of NiVpp, we engineered a hyperfusogenic NiV-F protein lacking an N-linked glycosylation site (T5FΔN3). T5FΔN3/wt G particles exhibited enhanced infectivity on less permissive cell lines and efficiently targeted ephrinB2(+) cells even in a 1,000-fold excess of ephrinB2-negative cells, all without any loss of specificity, as entry was abrogated by soluble ephrinB2. NiVpp also transduced human embryonic, hematopoietic, and neural stem cell populations in an ephrinB2-dependent manner. Finally, intravenous administration of the luciferase reporter NiVpp-T5FΔN3/G to mice resulted in signals being detected in the spleen and lung but not in the liver. Bypassing the liver sink is a critical barrier for targeted gene therapy. The extraordinary specificity of NiV-G for ephrinB2 holds promise for targeting specific ephrinB2(+) populations in vivo or in vitro.


Subject(s)
Ephrin-B2/metabolism , Genetic Vectors , Lentivirus/genetics , Nipah Virus/physiology , Receptors, Virus/metabolism , Stem Cells/virology , Virus Internalization , Animals , Cells, Cultured , Humans , Mice , Molecular Biology/methods , Nipah Virus/genetics , Transduction, Genetic
16.
Brain Behav Immun ; 26(4): 635-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22306453

ABSTRACT

Clinical studies suggest that stress-related biobehavioral factors can accelerate the progression of hematopoietic cancers such as acute lymphoblastic leukemia (ALL), but it is unclear whether such effects are causal or what biological pathways mediate such effects. Given the network of sympathetic nervous system (SNS) fibers that innervates the bone marrow to regulate normal (non-leukemic) hematopoietic progenitor cells, we tested the possibility that stress-induced SNS signaling might also affect ALL progression. In an orthotopic mouse model, Nalm-6 human pre-B ALL cells were transduced with the luciferase gene for longitudinal bioluminescent imaging and injected i.v. into male SCID mice for bone marrow engraftment. Two weeks of daily restraint stress significantly enhanced ALL tumor burden and dissemination in comparison to controls, and this effect was blocked by the ß-adrenergic antagonist, propranolol. Although Nalm-6 ALL cells expressed mRNA for ß1- and ß3-adrenergic receptors, they showed no evidence of cAMP signaling in response to norepinephrine, and norepinephrine failed to enhance Nalm-6 proliferation in vitro. These results show that chronic stress can accelerate the progression of human pre-B ALL tumor load via a ß-adrenergic signaling pathway that likely involves indirect regulation of ALL biology via alterations in the function of other host cell types such as immune cells or the bone marrow microenvironment.


Subject(s)
Leukemia, Experimental/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-3/metabolism , Stress, Psychological/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Disease Models, Animal , Disease Progression , Humans , Leukemia, Experimental/psychology , Male , Mice , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/psychology , Propranolol/pharmacology , Restraint, Physical , Signal Transduction/drug effects , Stress, Psychological/immunology
17.
Curr Opin Virol ; 1(1): 13-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21804908

ABSTRACT

Envelope virus replication begins with receptor binding, followed by fusion of the viral envelope with the cell membrane. The binding and fusion steps are usually mediated by envelope proteins. The ability of envelope proteins of a particular virus to bind and fuse with target cells defines the host range of the virus, known as 'viral tropism'. The mechanism(s) of fusion by the viral envelope is largely categorized as either pH-dependent or pH-independent. By redirecting the binding specificities of envelope proteins to desired target molecules while maintaining fusion activity, it is possible to redirect the tropisms of virus and viral vectors, enabling specific killing and/or transduction of desired cells in vivo. Recently, a lipid, phosphatidylserine, was also shown to mediate binding of virus, which affects the tropisms of viruses and viral vectors.


Subject(s)
Viral Envelope Proteins/metabolism , Viral Tropism , Virus Physiological Phenomena , Animals , Humans , Receptors, Virus/genetics , Receptors, Virus/metabolism , Viral Envelope Proteins/genetics , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/virology , Virus Internalization , Viruses/genetics
18.
Cell Host Microbe ; 9(4): 286-98, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21501828

ABSTRACT

Virus entry into cells is typically initiated by binding of virally encoded envelope proteins to specific cell surface receptors. Studying infectivity of lentivirus pseudotypes lacking envelope binding, we still observed high infectivity for some cell types. On further investigation, we discovered that this infectivity is conferred by the soluble bovine protein S in fetal calf serum, or Gas6, its human homolog. Gas6 enhances native infectivity of pseudotypes of multiple viral envelope proteins. Gas6 mediates binding of the virus to target cells by bridging virion envelope phosphatidylserine to Axl, a TAM receptor tyrosine kinase on target cells. Phagocytic clearance of apoptotic cells is known to involve bridging by Gas6. Replication of vaccinia virus, which was previously reported to use apoptotic mimicry to enter cells, is also enhanced by Gas6. These results reveal an alternative molecular mechanism of viral entry that can broaden host range and enhance infectivity of enveloped viruses.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Phosphatidylserines/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Humans , Phagocytosis/physiology , Protein S/metabolism , Receptors, Cell Surface , Sequence Homology, Amino Acid , Vaccinia virus/physiology , Virus Attachment , Axl Receptor Tyrosine Kinase
19.
Cancer Res ; 70(18): 7042-52, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20823155

ABSTRACT

Metastasis to distant tissues is the chief driver of breast cancer-related mortality, but little is known about the systemic physiologic dynamics that regulate this process. To investigate the role of neuroendocrine activation in cancer progression, we used in vivo bioluminescence imaging to track the development of metastasis in an orthotopic mouse model of breast cancer. Stress-induced neuroendocrine activation had a negligible effect on growth of the primary tumor but induced a 30-fold increase in metastasis to distant tissues including the lymph nodes and lung. These effects were mediated by ß-adrenergic signaling, which increased the infiltration of CD11b(+)F4/80(+) macrophages into primary tumor parenchyma and thereby induced a prometastatic gene expression signature accompanied by indications of M2 macrophage differentiation. Pharmacologic activation of ß-adrenergic signaling induced similar effects, and treatment of stressed animals with the ß-antagonist propranolol reversed the stress-induced macrophage infiltration and inhibited tumor spread to distant tissues. The effects of stress on distant metastasis were also inhibited by in vivo macrophage suppression using the CSF-1 receptor kinase inhibitor GW2580. These findings identify activation of the sympathetic nervous system as a novel neural regulator of breast cancer metastasis and suggest new strategies for antimetastatic therapies that target the ß-adrenergic induction of prometastatic gene expression in primary breast cancers.


Subject(s)
Adenocarcinoma/pathology , Mammary Neoplasms, Experimental/pathology , Stress, Physiological/physiology , Sympathetic Nervous System/pathology , Adenocarcinoma/immunology , Animals , Cell Line, Tumor , Female , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Stress, Physiological/immunology , T-Lymphocytes/immunology
20.
J Virol ; 84(14): 6923-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20484510

ABSTRACT

Redirecting the tropism of viral vectors enables specific transduction of selected cells by direct administration of vectors. We previously developed targeting lentiviral vectors by pseudotyping with modified Sindbis virus envelope proteins. These modified Sindbis virus envelope proteins have mutations in their original receptor-binding regions to eliminate their natural tropisms, and they are conjugated with targeting proteins, including antibodies and peptides, to confer their tropisms on target cells. We investigated whether our targeting vectors interact with DC-SIGN, which traps many types of viruses and gene therapy vectors by binding to the N-glycans of their envelope proteins. We found that these vectors do not interact with DC-SIGN. When these vectors were produced in the presence of deoxymannojirimycin, which alters the structures of N-glycans from complex to high mannose, these vectors used DC-SIGN as their receptor. Genetic analysis demonstrated that the N-glycans at E2 amino acid (aa) 196 and E1 aa 139 mediate binding to DC-SIGN, which supports the results of a previous report of cryoelectron microscopy analysis. In addition, we investigated whether modification of the N-glycan structures could activate serum complement activity, possibly by the lectin pathway of complement activation. DC-SIGN-targeted transduction occurs in the presence of human serum complement, demonstrating that high-mannose structure N-glycans of the envelope proteins do not activate human serum complement. These results indicate that the strategy of redirecting viral vectors according to alterations of their N-glycan structures would enable the vectors to target specific cells types expressing particular types of lectins.


Subject(s)
Cell Adhesion Molecules/metabolism , Genetic Vectors , Lectins, C-Type/metabolism , Lentivirus , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Sindbis Virus/metabolism , Viral Envelope Proteins , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Cell Adhesion Molecules/genetics , Cell Line , Complement System Proteins/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Lectins, C-Type/genetics , Lentivirus/genetics , Lentivirus/metabolism , Molecular Sequence Data , Polysaccharides/chemistry , Polysaccharides/genetics , Receptors, Cell Surface/genetics , Sindbis Virus/genetics , Transduction, Genetic , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...