Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Mol Cancer ; 17(1): 8, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343260

ABSTRACT

A vast array of tumor-derived genetic, proteomic and cellular components are constantly released into the circulation of cancer patients. These molecules including circulating tumor DNA and RNA, proteins, tumor and immune cells are emerging as convenient and accurate liquid biomarkers of cancer. Circulating cancer biomarkers provide invaluable information on cancer detection and diagnosis, prognosticate patient outcomes, and predict treatment response. In this era of effective molecular targeted treatments and immunotherapies, there is now an urgent need to implement use of these circulating biomarkers in the clinic to facilitate personalized therapy. In this review, we present recent findings in circulating melanoma biomarkers, examine the challenges and promise of evolving technologies used for liquid biomarker discovery, and discuss future directions and perspectives in melanoma biomarker research.


Subject(s)
Biomarkers, Tumor , Melanoma/diagnosis , Melanoma/metabolism , Animals , Clinical Trials as Topic , Early Detection of Cancer , Humans , Immunotherapy , Liquid Biopsy , Melanoma/therapy , Molecular Targeted Therapy , Proteomics/methods
2.
J Virol ; 90(4): 2102-11, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26656703

ABSTRACT

UNLABELLED: The alphaherpesviral envelope protein pUS9 has been shown to play a role in the anterograde axonal transport of herpes simplex virus 1 (HSV-1), yet the molecular mechanism is unknown. To address this, we used an in vitro pulldown assay to define a series of five arginine residues within the conserved pUS9 basic domain that were essential for binding the molecular motor kinesin-1. The mutation of these pUS9 arginine residues to asparagine blocked the binding of both recombinant and native kinesin-1. We next generated HSV-1 with the same pUS9 arginine residues mutated to asparagine (HSV-1pUS9KBDM) and then restored them being to arginine (HSV-1pUS9KBDR). The two mutated viruses were analyzed initially in a zosteriform model of recurrent cutaneous infection. The primary skin lesion scores were identical in severity and kinetics, and there were no differences in viral load at dorsal root ganglionic (DRG) neurons at day 4 postinfection (p.i.) for both viruses. In contrast, HSV-1pUS9KBDM showed a partial reduction in secondary skin lesions at day 8 p.i. compared to the level for HSV-1pUS9KBDR. The use of rat DRG neuronal cultures in a microfluidic chamber system showed both a reduction in anterograde axonal transport and spread from axons to nonneuronal cells for HSV-1pUS9KBDM. Therefore, the basic domain of pUS9 contributes to anterograde axonal transport and spread of HSV-1 from neurons to the skin through recruitment of kinesin-1. IMPORTANCE: Herpes simplex virus 1 and 2 cause genital herpes, blindness, encephalitis, and occasionally neonatal deaths. There is also increasing evidence that sexually transmitted genital herpes increases HIV acquisition, and the reactivation of HSV increases HIV replication and transmission. New antiviral strategies are required to control resistant viruses and to block HSV spread, thereby reducing HIV acquisition and transmission. These aims will be facilitated through understanding how HSV is transported down nerves and into skin. In this study, we have defined how a key viral protein plays a role in both axonal transport and spread of the virus from nerve cells to the skin.


Subject(s)
Axonal Transport , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Kinesins/metabolism , Lipoproteins/metabolism , Neurons/virology , Phosphoproteins/metabolism , Viral Proteins/metabolism , Virus Release , Amino Acid Sequence , Animals , Binding Sites , Cytological Techniques , DNA Mutational Analysis , Disease Models, Animal , Female , Ganglia, Spinal/virology , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins/genetics , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Protein Binding , Protein Interaction Mapping , Rats, Wistar , Severity of Illness Index , Skin/pathology , Skin/virology , Viral Load , Viral Proteins/genetics
3.
Antimicrob Agents Chemother ; 60(2): 1003-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643336

ABSTRACT

A marine-derived compound, abalone hemocyanin, from Haliotis rubra was shown to have a unique mechanism of antiviral activity against herpes simplex virus 1 (HSV-1) infections. In vitro assays demonstrated the dose-dependent and inhibitory effect of purified hemocyanin against HSV-1 infection in Vero cells with a 50% effective dose (ED50) of 40 to 50 nM and no significant toxicity. In addition, hemocyanin specifically inhibited viral attachment and entry by binding selectively to the viral surface glycoproteins gD, gB, and gC, probably by mimicking their receptors. However, hemocyanin had no effect on postentry events and did not block infection by binding to cellular receptors for HSV. By the use of different mutants of gD and gB and a competitive heparin binding assay, both protein charge and conformation were shown to be the driving forces of the interaction between hemocyanin and viral glycoproteins. These findings also suggested that hemocyanin may have different motifs for binding to each of the viral glycoproteins B and D. The dimer subunit of hemocyanin with a 10-fold-smaller molecular mass exhibited similar binding to viral surface glycoproteins, showing that the observed inhibition did not require the entire multimer. Therefore, a small hemocyanin analogue could serve as a new antiviral candidate for HSV infections.


Subject(s)
Antiviral Agents/pharmacology , Hemocyanins/pharmacology , Herpesvirus 1, Human/drug effects , Animals , Binding Sites , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gastropoda/chemistry , Glycoproteins/metabolism , Hemocyanins/isolation & purification , Hemocyanins/metabolism , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Vero Cells/drug effects , Vero Cells/virology
4.
Biochem Biophys Res Commun ; 470(3): 735-740, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26792716

ABSTRACT

Nuclear import of the accessory protein Vpr is central to infection by human immunodeficiency virus (HIV). We previously identified the Vpr F72L mutation in a HIV-infected, long-term non-progressor, showing that it resulted in reduced Vpr nuclear accumulation and altered cytoplasmic localisation. Here we demonstrate for the first time that the effects of nuclear accumulation of the F72L mutation are due to impairment of microtubule-dependent-enhancement of Vpr nuclear import. We use high resolution imaging approaches including fluorescence recovery after photobleaching and other approaches to document interaction between Vpr and the dynein light chain protein, DYNLT1, and impaired interaction of the F72L mutant with DYNLT1. The results implicate MTs/DYNLT1 as drivers of Vpr nuclear import and HIV infection, with important therapeutic implications.


Subject(s)
Cell Nucleus/metabolism , Dyneins/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Protein Binding , Protein Transport/physiology , Structure-Activity Relationship
5.
J Virol ; 90(5): 2653-63, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26699637

ABSTRACT

UNLABELLED: The herpes simplex virus type 1 (HSV-1) envelope protein pUS9 plays an important role in virus anterograde axonal transport and spread from neuronal axons. In this study, we used both confocal microscopy and transmission electron microscopy (TEM) to examine the role of pUS9 in the anterograde transport and assembly of HSV-1 in the distal axon of human and rat dorsal root ganglion (DRG) neurons using US9 deletion (US9(-)), repair (US9R), and wild-type (strain F, 17, and KOS) viruses. Using confocal microscopy and single and trichamber culture systems, we observed a reduction but not complete block in the anterograde axonal transport of capsids to distal axons as well as a marked (∼90%) reduction in virus spread from axons to Vero cells with the US9 deletion viruses. Axonal transport of glycoproteins (gC, gD, and gE) was unaffected. Using TEM, there was a marked reduction or absence of enveloped capsids, in varicosities and growth cones, in KOS strain and US9 deletion viruses, respectively. Capsids (40 to 75%) in varicosities and growth cones infected with strain 17, F, and US9 repair viruses were fully enveloped compared to less than 5% of capsids found in distal axons infected with the KOS strain virus (which also lacks pUS9) and still lower (<2%) with the US9 deletion viruses. Hence, there was a secondary defect in virus assembly in distal axons in the absence of pUS9 despite the presence of key envelope proteins. Overall, our study supports a dual role for pUS9, first in anterograde axonal transport and second in virus assembly in growth cones in distal axons. IMPORTANCE: HSV-1 has evolved mechanisms for its efficient transport along sensory axons and subsequent spread from axons to epithelial cells after reactivation. In this study, we show that deletion of the envelope protein pUS9 leads to defects in virus transport along axons (partial defect) and in virus assembly and egress from growth cones (marked defect). Virus assembly and exit in the neuronal cell body are not impaired in the absence of pUS9. Thus, our findings indicate that pUS9 contributes to the overall HSV-1 anterograde axonal transport, including a major role in virus assembly at the axon terminus, which is not essential in the neuronal cell body. Overall, our data suggest that the process of virus assembly at the growth cones differs from that in the neuronal cell body and that HSV-1 has evolved different mechanisms for virus assembly and exit from different cellular compartments.


Subject(s)
Axonal Transport , Growth Cones/virology , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Lipoproteins/metabolism , Phosphoproteins/metabolism , Viral Proteins/metabolism , Virus Assembly , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/virology , Humans , Intracellular Signaling Peptides and Proteins , Microscopy, Confocal , Microscopy, Electron, Transmission , Organ Culture Techniques , Rats, Wistar
6.
J Virol ; 89(13): 6575-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25855743

ABSTRACT

UNLABELLED: Dendritic cells (DCs) and macrophages are present in the tissues of the anogenital tract, where HIV-1 transmission occurs in almost all cases. These cells are both target cells for HIV-1 and represent the first opportunity for the virus to interfere with innate recognition. Previously we have shown that both cell types fail to produce type I interferons (IFNs) in response to HIV-1 but that, unlike T cells, the virus does not block IFN induction by targeting IFN regulatory factor 3 (IRF3) for cellular degradation. Thus, either HIV-1 inhibits IFN induction by an alternate mechanism or, less likely, these cells fail to sense HIV-1. Here we show that HIV-1 (but not herpes simplex virus 2 [HSV-2] or Sendai virus)-exposed DCs and macrophages fail to induce the expression of all known type I and III IFN genes. These cells do sense the virus, and pattern recognition receptor (PRR)-induced signaling pathways are triggered. The precise stage in the IFN-inducing signaling pathway that HIV-1 targets to block IFN induction was identified; phosphorylation but not K63 polyubiquitination of TANK-binding kinase 1 (TBK1) was completely inhibited. Two HIV-1 accessory proteins, Vpr and Vif, were shown to bind to TBK1, and their individual deletion partly restored IFN-ß expression. Thus, the inhibition of TBK1 autophosphorylation by binding of these proteins appears to be the principal mechanism by which HIV-1 blocks type I and III IFN induction in myeloid cells. IMPORTANCE: Dendritic cells (DCs) and macrophages are key HIV target cells. Therefore, definition of how HIV impairs innate immune responses to initially establish infection is essential to design preventative interventions, especially by restoring initial interferon production. Here we demonstrate how HIV-1 blocks interferon induction by inhibiting the function of a key kinase in the interferon signaling pathway, TBK1, via two different viral accessory proteins. Other viral proteins have been shown to target the general effects of TBK1, but this precise targeting between ubiquitination and phosphorylation of TBK1 is novel.


Subject(s)
Dendritic Cells/immunology , HIV-1/immunology , Host-Pathogen Interactions , Macrophages/immunology , Protein Serine-Threonine Kinases/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Dendritic Cells/virology , Humans , Immune Evasion , Interferons/antagonists & inhibitors , Macrophages/virology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Ubiquitination
7.
PLoS Pathog ; 9(3): e1003239, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555252

ABSTRACT

Cell-to-cell transmission of vaccinia virus can be mediated by enveloped virions that remain attached to the outer surface of the cell or those released into the medium. During egress, the outer membrane of the double-enveloped virus fuses with the plasma membrane leaving extracellular virus attached to the cell surface via viral envelope proteins. Here we report that F-actin nucleation by the viral protein A36 promotes the disengagement of virus attachment and release of enveloped virus. Cells infected with the A36(YdF) virus, which has mutations at two critical tyrosine residues abrogating localised actin nucleation, displayed a 10-fold reduction in virus release. We examined A36(YdF) infected cells by transmission electron microscopy and observed that during release, virus appeared trapped in small invaginations at the plasma membrane. To further characterise the mechanism by which actin nucleation drives the dissociation of enveloped virus from the cell surface, we examined recombinant viruses by super-resolution microscopy. Fluorescently-tagged A36 was visualised at sub-viral resolution to image cell-virus attachment in mutant and parental backgrounds. We confirmed that A36(YdF) extracellular virus remained closely associated to the plasma membrane in small membrane pits. Virus-induced actin nucleation reduced the extent of association, thereby promoting the untethering of virus from the cell surface. Virus release can be enhanced via a point mutation in the luminal region of B5 (P189S), another virus envelope protein. We found that the B5(P189S) mutation led to reduced contact between extracellular virus and the host membrane during release, even in the absence of virus-induced actin nucleation. Our results posit that during release virus is tightly tethered to the host cell through interactions mediated by viral envelope proteins. Untethering of virus into the surrounding extracellular space requires these interactions be relieved, either through the force of actin nucleation or by mutations in luminal proteins that weaken these interactions.


Subject(s)
Actin Cytoskeleton/metabolism , Vaccinia virus/physiology , Vaccinia/transmission , Viral Envelope Proteins/metabolism , Viral Structural Proteins/metabolism , Virus Release/physiology , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Membrane/ultrastructure , Cell Membrane/virology , Chlorocebus aethiops , Comet Assay , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fibroblasts/virology , Host-Pathogen Interactions , Mice , Microscopy, Electron, Transmission , NIH 3T3 Cells , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Vaccinia virus/ultrastructure , Vero Cells , Viral Envelope Proteins/ultrastructure , Viral Structural Proteins/ultrastructure
8.
J Virol ; 86(13): 7427-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532690

ABSTRACT

Egress of wrapped virus (WV) to the cell periphery following vaccinia virus (VACV) replication is dependent on interactions with the microtubule motor complex kinesin-1 and is mediated by the viral envelope protein A36. Here we report that ectromelia virus (ECTV), a related orthopoxvirus and the causative agent of mousepox, encodes an A36 homologue (ECTV-Mos-142) that is highly conserved despite a large truncation at the C terminus. Deleting the ECTV A36R gene leads to a reduction in the number of extracellular viruses formed and to a reduced plaque size, consistent with a role in microtubule transport. We also observed a complete loss of virus-associated actin comets, another phenotype dependent on A36 expression during VACV infection. ECTV ΔA36R was severely attenuated when used to infect the normally susceptible BALB/c mouse strain. ECTV ΔA36R replication and spread from the draining lymph nodes to the liver and spleen were significantly reduced in BALB/c mice and in Rag-1-deficient mice, which lack T and B lymphocytes. The dramatic reduction in ECTV ΔA36R titers early during the course of infection was not associated with an augmented immune response. Taken together, these findings demonstrate the critical role that subcellular transport pathways play not only in orthopoxvirus infection in an in vitro context but also during orthopoxvirus pathogenesis in a natural host. Furthermore, despite the attenuation of the mutant virus, we found that infection nonetheless induced protective immunity in mice, suggesting that orthopoxvirus vectors with A36 deletions may be considered another safe vaccine alternative.


Subject(s)
Cytoskeletal Proteins/metabolism , Ectromelia virus/pathogenicity , Ectromelia, Infectious/virology , Host-Pathogen Interactions , Viral Proteins/metabolism , Virus Release , Animals , Ectromelia virus/genetics , Female , Gene Deletion , Liver/virology , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Protein Transport , Spleen/virology , Viral Load , Viral Plaque Assay , Viral Proteins/genetics , Virulence
9.
J Virol ; 86(11): 6123-37, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22457528

ABSTRACT

Herpes simplex virus 1 (HSV-1) enters neurons primarily by fusion of the viral envelope with the host cell plasma membrane, leading to the release of the capsid into the cytosol. The capsid travels via microtubule-mediated retrograde transport to the nuclear membrane, where the viral DNA is released for replication in the nucleus. In the present study, the composition and kinetics of incoming HSV-1 capsids during entry and retrograde transport in axons of human fetal and dissociated rat dorsal root ganglia (DRG) neurons were examined by wide-field deconvolution microscopy and transmission immunoelectron microscopy (TIEM). We show that HSV-1 tegument proteins, including VP16, VP22, most pUL37, and some pUL36, dissociated from the incoming virions. The inner tegument proteins, including pUL36 and some pUL37, remained associated with the capsid during virus entry and transit to the nucleus in the neuronal cell body. By TIEM, a progressive loss of tegument proteins, including VP16, VP22, most pUL37, and some pUL36, was observed, with most of the tegument dissociating at the plasma membrane of the axons and the neuronal cell body. Further dissociation occurred within the axons and the cytosol as the capsids moved to the nucleus, resulting in the release of free tegument proteins, especially VP16, VP22, pUL37, and some pUL36, into the cytosol. This study elucidates ultrastructurally the composition of HSV-1 capsids that encounter the microtubules in the core of human axons and the complement of free tegument proteins released into the cytosol during virus entry.


Subject(s)
Ganglia, Spinal/virology , Herpesvirus 1, Human/physiology , Neurons/virology , Viral Structural Proteins/metabolism , Virus Internalization , Animals , Cells, Cultured , Herpesvirus 1, Human/ultrastructure , Humans , Microscopy , Rats , Rats, Wistar , Viral Structural Proteins/ultrastructure , Virion/ultrastructure
10.
Biochem Biophys Res Commun ; 417(3): 961-5, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22202175

ABSTRACT

The herpes simplex virus type 1 (HSV-1) structural tegument protein pUL37, which is conserved across the Herpesviridae family, is known to be essential for secondary envelopment during the egress of viral particles. To shed light on additional roles of pUL37 during viral replication a yeast two-hybrid screen of a human brain cDNA library was undertaken. This screen identified ten host cell proteins as potential pUL37 interactors. One of the interactors, serine threonine kinase TAOK3, was subsequently confirmed to interact with pUL37 using an in vitro pulldown assay. Such host cell/pUL37 interactions provide further insights into the multifunctional role of this herpesviral tegument protein.


Subject(s)
Brain/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Protein Serine-Threonine Kinases/metabolism , Viral Structural Proteins/metabolism , Brain/virology , Gene Library , Herpesvirus 1, Human/metabolism , Humans , Two-Hybrid System Techniques , Virus Replication
11.
Cancer Gene Ther ; 29(10): 1384-1393, 2022 10.
Article in English | MEDLINE | ID: mdl-35352024

ABSTRACT

Uveal melanoma (UM) is a rare cancer arising from melanocytes in the uveal tract of the eye. Despite effective primary treatment, there is no approved therapy for metastatic UM and prognosis and survival remain poor. Over 90% of UM are driven by mutations affecting the Gα subunits encoded by the GNAQ and GNA11 genes. These mutations activate downstream and targetable signaling pathways, including the protein kinase C (PKC) cascade. PKC inhibitors have been used in clinical trials for metastatic UM but have shown limited efficacy. In this study, we examined the signaling and functional effects of two PKC inhibitors (AEB071 and IDE196) in a panel of UM cell models. In response to PKC inhibition, all UM cell lines showed potent suppression of PKC activity, but this was not sufficient to predict PKC inhibitor sensitivity and only two UM cell lines showed substantial PKC inhibitor-induced cell death. The differences in UM cell responses to PKC inhibition were not attributable to the degree or timing of PKC suppression or inhibition of the downstream mitogen-activated protein kinase (MAPK) or phosphatidylinositol-3-kinase (PI3K) pathways. Instead, UM cell show complex, PKC-independent signaling pathways that contribute to their survival and resistance to targeted therapies.


Subject(s)
Protein Kinase Inhibitors , Uveal Neoplasms , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/therapeutic use , Humans , Melanoma , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositols/therapeutic use , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
12.
Am J Surg Pathol ; 46(8): 1048-1059, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35439782

ABSTRACT

Blue nevi are benign, melanocytic neoplasms that show a range of clinical and morphologic patterns and include common/dendritic, cellular, and atypical cellular subtypes. Like other nevi, they most commonly occur in skin but can occasionally involve lymph nodes where they may be misinterpreted as representing metastatic melanoma. Moreover, whether benign blue nevi can metastasize to lymph nodes and their natural history and prognostic significance has been the subject of great controversy. To date, few cases of nodal blue nevi have been reported in the literature, and those reports have had limited clinical follow-up and supporting molecular data. This study sought to determine the clinical, pathologic, and molecular features of blue nevi involving lymph nodes, clarify their clinical significance, provide evidence for understanding their pathogenesis, and highlight potential pitfalls in the interpretation of lymph nodes with an ultimate aim of improving patient care. Thirteen cases of blue nevi involving lymph nodes were identified in the archives of Royal Prince Alfred Hospital, Sydney, Australia (1984-2018). A detailed assessment of the clinical and pathologic features of each case was performed, including an evaluation of all available immunohistochemical stains. Extended clinical follow-up was available for 9 patients. Droplet digital polymerase chain reaction for GNAQ Q209L, Q209P and GNA11 Q209L mutations was performed on 7 cases of blue nevi within lymph nodes together with matching cutaneous (presumed primary) blue nevi in 2 cases. All cases showed typical histologic features of blue nevi. BAP1 was retained in all cases (n=7). There were no recurrence or metastasis of blue nevus in any case on long-term clinical follow-up (n=9, median follow-up, 12 y). The majority of cases (n=5 of 7 evaluated) had GNAQ and GNA11 driver mutations. The 2 patients with a matched primary cutaneous blue nevus and regionally associated nodal blue nevus had the same GNAQ Q209L mutation in both sites in each patient. We conclude that blue nevi can involve lymph nodes and are associated with benign clinical behavior, and probably represent so-called "benign" metastasis. Awareness of these lesions is important when evaluating lymph nodes to avoid misdiagnosis as metastatic melanoma.


Subject(s)
Melanoma , Nevus, Blue , Nevus , Skin Neoplasms , Follow-Up Studies , Humans , Melanoma/pathology , Nevus/pathology , Nevus, Blue/genetics , Nevus, Blue/pathology , Skin Neoplasms/pathology
13.
Front Oncol ; 12: 820510, 2022.
Article in English | MEDLINE | ID: mdl-35494035

ABSTRACT

Detection of melanoma mutations using circulating tumor DNA (ctDNA) is a potential alternative to using genomic DNA from invasive tissue biopsies. To date, mutations in the GC-rich TERT promoter region, which is commonly mutated in melanoma, have been technically difficult to detect in ctDNA using next-generation sequencing (NGS) panels. In this study, we developed a custom melanoma NGS panel for detection of ctDNA, which encompasses the top 15 gene mutations in melanoma including the TERT promoter. We analyzed 21 stage III and IV melanoma patient samples who were treatment-naïve or on therapy. The overall detection rate of the custom panel, based on BRAF/NRAS/TERT promoter mutations, was 14/21 (67%) patient samples which included a TERT C250T mutation in one BRAF and NRAS mutation negative sample. A BRAF or NRAS mutation was detected in the ctDNA of 13/21 (62%) patients while TERT promoter mutations were detected in 10/21 (48%) patients. Co-occurrence of TERT promoter mutations with BRAF or NRAS mutations was found in 9/10 (90%) patients. The custom ctDNA panel showed a concordance of 16/21 (76%) with tissue based-detection and included 12 BRAF/NRAS mutation positive and 4 BRAF/NRAS mutation negative patients. The ctDNA mutation detection rate for stage IV was 12/16 (75%) and for stage III was 1/5 (20%). Based on BRAF, NRAS and TERT promoter mutations, the custom melanoma panel displayed a limit of detection of ~0.2% mutant allele frequency and showed significant correlation with droplet digital PCR. For one patient, a novel MAP2K1 H119Y mutation was detected in an NRAS/BRAF/TERT promoter mutation negative background. To increase the detection rate to >90% for stage IV melanoma patients, we plan to expand our custom panel to 50 genes. This study represents one of the first to successfully detect TERT promoter mutations in ctDNA from cutaneous melanoma patients using a targeted NGS panel.

14.
J Virol ; 84(3): 1397-405, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923173

ABSTRACT

In this study a number of herpes simplex virus type 1 (HSV-1) proteins were screened, using a yeast-two-hybrid assay, for interaction with the tegument protein pUL48 (VP16). This approach identified interactions between pUL48 and the capsid proteins pUL19 (VP5), pUL38 (VP19C), and pUL35 (VP26). In addition, the previously identified interaction of pUL48 with the major tegument protein pUL36 (VP1/2) was confirmed. All of these interactions, except that of pUL35 and pUL48, could be confirmed using an in vitro pulldown assay. A subsequent pulldown assay with intact in vitro-assembled capsids, consisting of pUL19, pUL38, and pUL18 (VP23) with or without pUL35, showed no binding of pUL48, suggesting that the capsid/pUL48 interactions initially identified were more then likely not biologically relevant. This was confirmed by using a series of HSV-1 mutants lacking the gene encoding either pUL35, pUL36, or pUL37. For each HSV-1 mutant, analysis of purified deenveloped C capsids indicated that only in the absence of pUL36 was there a complete loss of capsid-bound pUL48, as well as pUL37. Collectively, this study shows for the first time that pUL36 is a major factor for addition of both pUL48 and pUL37, likely through a direct interaction of both with nonoverlapping sites in pUL36, to unenveloped C capsids during assembly of HSV-1.


Subject(s)
Capsid , Herpes Simplex Virus Protein Vmw65/physiology , Herpesvirus 1, Human/physiology , Viral Proteins/physiology , Animals , Chlorocebus aethiops , HeLa Cells , Humans , Two-Hybrid System Techniques , Vero Cells
15.
Access Microbiol ; 3(3): 000206, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34151161

ABSTRACT

HSV-1 envelope glycoprotein E (gE) is important for viral egress and cell-to-cell spread but the host protein(s) involved in these functions have yet to be determined. We aimed to investigate a role for the Arp2/3 complex and actin regulation in viral egress based on the identification of a WAVE Regulatory Complex (WRC) Interacting Receptor Sequence (WIRS) in the cytoplasmic tail (CT) of gE. A WIRS-dependent interaction between the gE(CT) and subunits of the WRC was demonstrated by GST-pulldown assay and a role for the Arp2/3 complex in cell-to-cell spread was also observed by plaque assay. Subsequent study of a recombinant HSV-1 gE WIRS-mutant found no significant changes to viral production and release based on growth kinetics studies, or changes to plaque and comet size in various cell types, suggesting no function for the motif in cell-to-cell spread. GFP-Trap pulldown and proximity ligation assays were unable to confirm a WIRS-dependent interaction between gE and the WRC in human cell lines though the WIRS-independent interaction observed in situ warrants further study. Confocal microscopy of infected cells of neuronal origin identified no impairment of gE WIRS-mutant HSV-1 anterograde transport along axons. We propose that the identified gE WIRS motif does not function directly in recruitment of the WRC in human cells, in cell-to-cell spread of virus or in anterograde transport along axons. Further studies are needed to understand how HSV-1 manipulates and traverses the actin cytoskeleton and how gE may contribute to these processes in a WIRS-independent manner.

16.
Cancers (Basel) ; 13(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917514

ABSTRACT

The prognosis for patients with UM is poor, and recent clinical trials have failed to prolong overall survival (OS) of these patients. Over 95% of UM harbor activating driver mutations, and this allows for the investigation of ctDNA. In this study, we investigated the value of ctDNA for adaptive clinical trial design in metastatic UM. Longitudinal plasma samples were analyzed for ctDNA in 17 metastatic UM patients treated with PKCi-based therapy in a phase 1 clinical trial setting. Plasma ctDNA was assessed using digital droplet PCR (ddPCR) and a custom melanoma gene panel for targeted next generation sequencing (NGS). Baseline ctDNA strongly correlated with baseline lactate dehydrogenase (LDH) (p < 0.001) and baseline disease burden (p = 0.002). Early during treatment (EDT) ctDNA accurately predicted patients with clinical benefit to PKCi using receiver operator characteristic (ROC) curves (AUC 0.84, [95% confidence interval 0.65-1.0, p = 0.026]). Longitudinal ctDNA assessment was informative for establishing clinical benefit and detecting disease progression with 7/8 (88%) of patients showing a rise in ctDNA and targeted NGS of ctDNA revealed putative resistance mechanisms prior to radiological progression. The inclusion of longitudinal ctDNA monitoring in metastatic UM can advance adaptive clinical trial design.

17.
J Gen Virol ; 91(Pt 11): 2659-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20668116

ABSTRACT

In this study, fragments of the small capsid protein pUL35 (VP26) from herpes simplex virus type 1 (HSV-1) were generated to identify binding domains for a number of known ligands. Analysis of the binding of dynein light chain subunits, DYNLT1 and DYNLT3, as well the HSV-1 structural proteins pUL19 (VP5) and pUL37 was then undertaken using the LexA yeast two-hybrid assay. The N-terminal half of pUL35, in particular residues 30-43, was identified as a common region for the binding of DYNLT1 and DYNLT3. Additional distinct regions in the C terminus of pUL35 also contribute to the binding of DYNLT1 and DYNLT3. In contrast, only the C-terminal half of pUL35 was found to mediate the binding of pUL19 and pUL37 through distinct regions. The relevance of this information to the role of pUL35 in viral transport and assembly is discussed.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/metabolism , Dyneins/metabolism , Herpesvirus 1, Human/physiology , Virus Assembly , Virus Replication , Binding Sites , Protein Binding , Protein Interaction Mapping , Two-Hybrid System Techniques , Viral Structural Proteins/metabolism
18.
Biochem Biophys Res Commun ; 391(1): 388-93, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19913510

ABSTRACT

The cellular molecular motor kinesin-1 mediates the microtubule-dependent transport of a range of cargo. We have previously identified an interaction between the cargo-binding domain of kinesin-1 heavy chain KIF5B and the membrane-associated SNARE proteins SNAP-25 and SNAP-23. In this study we further defined the minimal SNAP-25 binding domain in KIF5B to residues 874-894. Overexpression of a fragment of KIF5B (residues 594-910) resulted in significant colocalization with SNAP-25 with resulting blockage of the trafficking of SNAP-25 to the periphery of cells. This indicates that kinesin-1 facilitates the transport of SNAP-25 containing vesicles as a prerequisite to SNAP-25 driven membrane fusion events.


Subject(s)
Cell Membrane/metabolism , Kinesins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Binding Sites , HeLa Cells , Humans , Kinesins/genetics , Protein Structure, Tertiary , Protein Transport , Synaptic Vesicles/metabolism
19.
J Virol ; 83(7): 3187-99, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176621

ABSTRACT

Axonal transport of herpes simplex virus (HSV-1) is essential for viral infection and spread in the peripheral nervous system of the host. Therefore, the virus probably utilizes existing active transport and targeting mechanisms in neurons for virus assembly and spread from neurons to skin. In the present study, we used transmission immunoelectron microscopy to investigate the nature and origin of vesicles involved in the anterograde axonal transport of HSV-1 tegument and envelope proteins and of vesicles surrounding partially and fully enveloped capsids in growth cones. This study aimed to elucidate the mechanism of virus assembly and exit from axons of human fetal dorsal root ganglia neurons. We demonstrated that viral tegument and envelope proteins can travel in axons independently of viral capsids and were transported to the axon terminus in two types of transport vesicles, tubulovesicular membrane structures and large dense-cored vesicles. These vesicles and membrane carriers were derived from the trans-Golgi network (TGN) and contained key proteins, such as Rab3A, SNAP-25, GAP-43, and kinesin-1, involved in the secretory and exocytic pathways in axons. These proteins were also observed on fully and partially enveloped capsids in growth cones and on extracellular virions. Our findings provide further evidence to the subassembly model of separate transport in axons of unenveloped capsids from envelope and tegument proteins with final virus assembly occurring at the axon terminus. We postulate that HSV-1 capsids invaginate tegument- and envelope-bearing TGN-derived vesicles and utilize the large secretory vesicle pathway of exocytosis for exit from axons.


Subject(s)
Axons/virology , Exocytosis , Growth Cones/virology , Herpesvirus 1, Human/physiology , Secretory Vesicles/virology , Viral Structural Proteins/metabolism , Virus Assembly , Axons/ultrastructure , Cell Line , GAP-43 Protein/analysis , Growth Cones/ultrastructure , Humans , Kinesins/analysis , Microscopy, Immunoelectron , Secretory Vesicles/chemistry , Synaptosomal-Associated Protein 25/analysis , rab3A GTP-Binding Protein/analysis
20.
Methods Mol Biol ; 2060: 1-30, 2020.
Article in English | MEDLINE | ID: mdl-31617170

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is a prevalent and important human pathogen that has been studied in a wide variety of contexts. This book provides protocols currently in use in leading laboratories in many fields of HSV-1 research. This introductory chapter gives a brief overview of HSV-1 biology and life cycle, covering basic aspects of virus structure, the prevalence of and diseases caused by the virus, replication in cultured cells, viral latency, antiviral defenses, and the mechanisms that the virus uses to counteract these defenses.


Subject(s)
Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Virus Activation/physiology , Virus Latency/physiology , Animals , Cell Line , Humans
SELECTION OF CITATIONS
SEARCH DETAIL