Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 20(6): e1012267, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38857290

ABSTRACT

HSV infects keratinocytes in the epidermis of skin via nectin-1. We established a human foreskin explant infection model to investigate HSV entry and spread. HSV1 entry could only be achieved by the topical application of virus via high density microarray projections (HD-MAPs) to the epidermis, which penetrated beyond one third of its thickness, simulating in vivo microtrauma. Rapid lateral spread of HSV1 to a mean of 13 keratinocytes wide occurred after 24 hours and free virus particles were observed between keratinocytes, consistent with an intercellular route of spread. Nectin-1 staining was markedly decreased in foci of infection in the epidermis and in the human keratinocyte HaCaT cell line. Nectin-1 was redistributed, at the protein level, in adjacent uninfected cells surrounding infection, inducible by CCL3, IL-8 (or CXCL8), and possibly CXCL10 and IL-6, thus facilitating spread. These findings provide the first insights into HSV1 entry and spread in human inner foreskin in situ.


Subject(s)
Chemokines , Foreskin , Herpes Simplex , Herpesvirus 1, Human , Keratinocytes , Nectins , Humans , Male , Keratinocytes/virology , Keratinocytes/metabolism , Foreskin/virology , Foreskin/cytology , Nectins/metabolism , Herpes Simplex/virology , Herpes Simplex/metabolism , Chemokines/metabolism , Herpesvirus 1, Human/physiology , Cell Adhesion Molecules/metabolism , Virus Internalization
2.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055133

ABSTRACT

Herpesviruses are highly prevalent in the human population, and frequent reactivations occur throughout life. Despite antiviral drugs against herpetic infections, the increasing appearance of drug-resistant viral strains and their adverse effects prompt the research of novel antiherpetic drugs for treating lesions. Peptides obtained from natural sources have recently become of particular interest for antiviral therapy applications. In this work, we investigated the antiviral activity of the peptide A-3302-B, isolated from a marine bacterium, Micromonospora sp., strain MAG 9-7, against herpes simplex virus type 1, type 2, and human cytomegalovirus. Results showed that the peptide exerted a specific inhibitory activity against HSV-2 with an EC50 value of 14 µM. Specific antiviral assays were performed to investigate the mechanism of action of A-3302-B. We demonstrated that the peptide did not affect the expression of viral proteins, but it inhibited the late events of the HSV-2 replicative cycle. In detail, it reduced the cell-to-cell virus spread and the transmission of the extracellular free virus by preventing the egress of HSV-2 progeny from the infected cells. The dual antiviral and previously reported anti-inflammatory activities of A-3302-B, and its effect against an acyclovir-resistant HSV-2 strain are attractive features for developing a therapeutic to reduce the transmission of HSV-2 infections.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 2, Human/physiology , Micromonospora/chemistry , Peptides/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Foreskin/cytology , Foreskin/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/drug effects , Humans , Male , Molecular Structure , Peptides/chemistry , Peptides/isolation & purification , Vero Cells , Virus Release/drug effects
3.
BJU Int ; 127(4): 428-434, 2021 04.
Article in English | MEDLINE | ID: mdl-32870593

ABSTRACT

OBJECTIVES: To investigate the prevalence of pathological disease and spectrum of human papillomavirus (HPV) types among symptomatic foreskin tissue. PATIENTS AND METHODS: Consecutively excised symptomatic foreskins from 351 men were sent for histopathological evaluation. During the surgical procedure, a fresh biopsy was taken for HPV analysis by modified general primer polymerase chain reaction. A medical questionnaire regarding medication, smoking habits, number of lifetime sexual partners, former diseases and surgery performed on penis was completed by all participants. RESULTS: The most common clinical diagnosis and cause for circumcision was phimosis, seen in 85.2%. Histopathologically inflammatory dermatological conditions were present in 87% of the men. The most common histopathological diagnosis was lichen sclerosus (LS) observed among 58.7%. Notably, penile intraepithelial neoplasia (PeIN) was present in 2% without former clinical suspicion. Overall, HPV was detected in 17.1% of the men and 28 different HPV types were found. High-risk (HR) HPV types were identified in 9.1% and HPV16 was present in 2.3%. Current smoking increased the risk of HPV (crude odds ratio [OR] 2.8, confidence interval [CI] 1.4-5.6; P = 0.005). Having >15 lifetime sexual partners increased the risk of HPV (crude OR 2.6, 95% CI 1.4-5.1; P = 0.003) and when adjusted for current smoking the OR was substantially increased (OR 6.0, 95% CI CI 2.2-16.8; P < 0001). CONCLUSIONS: Histopathological evaluation of circumcised symptomatic foreskin revealed PeIN in 2% of the men without any clinical suspicion of malignancy and that treatable dermatological conditions were present in 87%, LS being the most common. HR-HPV types were present in 9%. Due to risk of malignant development both in PeIN and in inflammatory skin diseases we recommend sending all excised foreskins from patients with symptoms for histopathological evaluation as guidance for further clinical management.


Subject(s)
Alphapapillomavirus/isolation & purification , Carcinoma in Situ/virology , Foreskin/virology , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Penile Neoplasms/virology , Adult , Circumcision, Male , Cross-Sectional Studies , Humans , Male , Middle Aged , Prevalence , Sweden
4.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31189713

ABSTRACT

The development of a vaccine against human cytomegalovirus infection (HCMV) is a high-priority medical goal. The viral pentameric protein complex consisting of glycoprotein H (gH)/gL/UL128-131A (PC) is considered to be an important vaccine component. Its relevance to the induction of a protective antibody response is, however, still a matter of debate. We addressed this issue by using subviral dense bodies (DBs) of HCMV. DBs are exceptionally immunogenic. Laboratory HCMV strain DBs harbor important neutralizing antibody targets, like the glycoproteins B, H, L, M, and N, but they are devoid of the PC. To be able to directly compare the impact of the PC on the levels of neutralizing antibody (NT-abs) responses, a PC-positive variant of the HCMV laboratory strain Towne was established by bacterial artificial chromosome (BAC) mutagenesis (Towne-UL130rep). This strain synthesized PC-positive DBs upon infection of fibroblasts. These DBs were used in side-by-side immunizations with PC-negative Towne DBs. Mouse and rabbit sera were tested to address the impact of the PC on DB immunogenicity. The neutralizing antibody response to PC-positive DBs was superior to that of PC-negative DBs, as tested on fibroblasts, epithelial cells, and endothelial cells and for both animal species used. The experiments revealed the potential of the PC to enhance the antibody response against HCMV. Of particular interest was the finding that PC-positive DBs induced an antibody response that blocked the infection of fibroblasts by a PC-positive viral strain more efficiently than sera following immunizations with PC-negative particles.IMPORTANCE Infections with the human cytomegalovirus (HCMV) may cause severe and even life-threatening disease manifestations in newborns and immunosuppressed individuals. Several strategies for the development of a vaccine against this virus are currently pursued. A critical question in this respect refers to the antigenic composition of a successful vaccine. Using a subviral particle vaccine candidate, we show here that one protein complex of HCMV, termed the pentameric complex (PC), enhances the neutralizing antibody response against viral infection of different cell types. We further show for the first time that this not only relates to the infection of epithelial or endothelial cells; the presence of the PC in the particles also enhanced the neutralizing antibody response against the infection of fibroblasts by HCMV. Together, these findings argue in favor of including the PC in strategies for HCMV vaccine development.


Subject(s)
Antibodies, Neutralizing/metabolism , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Viral Envelope Proteins/immunology , Animals , Cells, Cultured , Cytomegalovirus Vaccines/immunology , Foreskin/cytology , Foreskin/virology , Human Umbilical Vein Endothelial Cells , Humans , Male , Membrane Glycoproteins/immunology , Mice , Multiprotein Complexes/immunology , Rabbits
5.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31189705

ABSTRACT

Epidemiological data confirm a much higher incidence of high-risk human papillomavirus 16 (HPV16)-mediated carcinogenesis of the cervical epithelium than for other target sites. In order to elucidate tissue-specific responses to virus infection, we compared gene expression changes induced by productive HPV16 infection of cervical, foreskin, and tonsil organotypic rafts. These rafts closely mimic persistent HPV16 infection, long before carcinogenesis sets in. The total number of gene expression changes varied considerably across the tissue types, with only 32 genes being regulated in common. Among them, we confirmed the Kelch-like family protein KLHL35 and the laminin-5 complex to be upregulated and downregulated, respectively, in all the three tissues. HPV16 infection induces upregulation of genes involved in cell cycle control, cell division, mitosis, DNA replication, and DNA damage repair in all the three tissues, indicative of a hyperproliferative environment. In the cervical and tonsil epithelium, we observe significant downregulation of genes involved in epidermis development, keratinocyte differentiation, and extracellular matrix organization. On the other hand, in HPV16-positive foreskin (HPV16 foreskin) tissue, several genes involved in interferon-mediated innate immunity, cytokine signaling, and cellular defenses were downregulated. Furthermore, pathway analysis and experimental validations identified important cellular pathways like STAT1 and transforming growth factor ß (TGF-ß) to be differentially regulated among the three tissue types. The differential modulation of important cellular pathways like TGF-ß1 and STAT1 can explain the sensitivity of tissues to HPV cancer progression.IMPORTANCE Although the high-risk human papillomavirus 16 infects anogenital and oropharyngeal sites, the cervical epithelium has a unique vulnerability to progression of cancer. Host responses during persistent infection and preneoplastic stages can shape the outcome of cancer progression in a tissue-dependent manner. Our study for the first time reports differential regulation of critical cellular functions and signaling pathways during productive HPV16 infection of cervical, foreskin, and tonsil tissues. While the virus induces hyperproliferation in infected cells, it downregulates epithelial differentiation, epidermal development, and innate immune responses, according to the tissue type. Modulation of these biological functions can determine virus fitness and pathogenesis and illuminate key cellular mechanisms that the virus employs to establish persistence and finally initiate disease progression.


Subject(s)
Cervix Uteri/virology , Foreskin/virology , Gene Expression Profiling/methods , Human papillomavirus 16/pathogenicity , Palatine Tonsil/virology , Papillomavirus Infections/genetics , Cell Differentiation , Cell Line, Tumor , Cervix Uteri/chemistry , Cervix Uteri/cytology , Female , Foreskin/chemistry , Foreskin/cytology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Human papillomavirus 16/genetics , Humans , Keratinocytes/chemistry , Keratinocytes/cytology , Keratinocytes/virology , Male , Microarray Analysis , Organ Specificity , Palatine Tonsil/chemistry , Palatine Tonsil/cytology , Papillomavirus Infections/virology , Signal Transduction , Virus Replication
6.
PLoS Pathog ; 13(2): e1006247, 2017 02.
Article in English | MEDLINE | ID: mdl-28241053

ABSTRACT

Oropharyngeal mucosal epithelia of fetuses/neonates/infants and the genital epithelia of adults play a critical role in HIV-1 mother-to-child transmission and sexual transmission of virus, respectively. To study the mechanisms of HIV-1 transmission through mucosal epithelium, we established polarized tonsil, cervical and foreskin epithelial cells. Analysis of HIV-1 transmission through epithelial cells showed that approximately 0.05% of initially inoculated virions transmigrated via epithelium. More than 90% of internalized virions were sequestered in the endosomes of epithelial cells, including multivesicular bodies (MVBs) and vacuoles. Intraepithelial HIV-1 remained infectious for 9 days without viral release. Release of sequestered intraepithelial HIV-1 was induced by the calcium ionophore ionomycin and by cytochalasin D, which increase intracellular calcium and disrupt the cortical actin of epithelial cells, respectively. Cocultivation of epithelial cells containing HIV-1 with activated peripheral blood mononuclear cells and CD4+ T lymphocytes led to the disruption of epithelial cortical actin and spread of virus from epithelial cells to lymphocytes. Treatment of epithelial cells with proinflammatory cytokines tumor necrosis factor-alpha and interferon gamma also induced reorganization of cortical actin and release of virus. Inhibition of MVB formation by small interfering RNA (siRNA)-mediated silencing of its critical protein hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) expression reduced viral sequestration in epithelial cells and its transmission from epithelial cells to lymphocytes by ~60-70%. Furthermore, inhibition of vacuole formation of epithelial cells by siRNA-inactivated rabankyrin-5 expression also significantly reduced HIV-1 sequestration in epithelial cells and spread of virus from epithelial cells to lymphocytes. Interaction of the intercellular adhesion molecule-1 of epithelial cells with the function-associated antigen-1 of lymphocytes was important for inducing the release of sequestered HIV-1 from epithelial cells and facilitating cell-to-cell spread of virus from epithelial cells to lymphocytes. This mechanism may serve as a pathway of HIV-1 mucosal transmission.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Epithelial Cells/virology , HIV Infections/transmission , Mucous Membrane/virology , Transcytosis/physiology , Blotting, Western , Cervix Uteri/virology , Coculture Techniques , Dendritic Cells/virology , Female , Fluorescent Antibody Technique , Foreskin/virology , HIV-1 , Humans , Leukocytes, Mononuclear/virology , Macrophages/virology , Male , Palatine Tonsil/virology
7.
PLoS Pathog ; 13(2): e1006163, 2017 02.
Article in English | MEDLINE | ID: mdl-28207890

ABSTRACT

Understanding early events of HIV transmission within mucosal tissues is vital for developing effective prevention strategies. Here, we report that primary stromal fibroblasts isolated from endometrium, cervix, foreskin, male urethra, and intestines significantly increase HIV infection of CD4+ T cells-by up to 37-fold for R5-tropic HIV and 100-fold for X4-tropic HIV-without themselves becoming infected. Fibroblasts were more efficient than dendritic cells at trans-infection and mediate this response in the absence of the DC-SIGN and Siglec-1 receptors. In comparison, mucosal epithelial cells secrete antivirals and inhibit HIV infection. These data suggest that breaches in the epithelium allow external or luminal HIV to escape an antiviral environment to access the infection-favorable environment of the stromal fibroblasts, and suggest that resident fibroblasts have a central, but previously unrecognized, role in HIV acquisition at mucosal sites. Inhibiting fibroblast-mediated enhancement of HIV infection should be considered as a novel prevention strategy.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Fibroblasts/cytology , HIV Infections/transmission , HIV-1/pathogenicity , Mucous Membrane/virology , Coculture Techniques , Endometrium/cytology , Endometrium/virology , Female , Flow Cytometry , Foreskin/cytology , Foreskin/virology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/virology , Male , Mucous Membrane/cytology , Oligonucleotide Array Sequence Analysis , Urethra/cytology , Urethra/virology
8.
BMC Infect Dis ; 19(1): 122, 2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30727957

ABSTRACT

BACKGROUND: Human papillomavirus (HPV) contributes to the development of cervical and oropharyngeal tumors. The increased incidence of HPV associated oropharyngeal tumors is lately being observed also in Polish population. The worldwide distribution of HPV varies and the studies rarely combine analysis of virus genotypes in both: genital and oropharyngeal locations. Therefore, in our study, we investigated HPV distribution in both anatomical sites of females with previous history of cervical cancer or with pre-cancerous lesion and their partners to establish the dominant types in Polish couples in genital and oropharyngeal areas, as they can be easily sexually transmitted. METHODS: The study group consisted of 197 females and their partners. Each female had current or previous cervical pathology and HPV detected in gynecological swab with the use of Anyplex™ II HPV28 Detection system. This system is based on multiplexed real time PCR and enables detection of 19 high-risk and 9 low-risk HPVs. RESULTS: Beside women, the virus was found in 114/197 of men in their foreskin swabs. Additionally, HPV was detected in oropharyngeal swabs of 39/197 female and 56/197 male participants. HPV 16/31/42/39/54 dominated in female and HPV 66/42/16/31/53 in male genital locations. The incidence of HPV in oropharynx was lower, top five genotypes included: HPV 6/39/42/35/16 in women compared to HPV 39/6/42/40/33 in men. HPV16 was the most frequently detected virus type, found in 70/197 examined cervical swabs. It was significantly more prevalent as single infection in females, previously treated for the cervical cancer (p = 0.035). Moreover, regular presence of low risk type 42 was noticeable in both sexes, in both kind of swabs. There was a trend observed towards its prevalence as single infectious agent in women with previous history of cervical cancer (p = 0.069). CONCLUSIONS: Our results showed the distribution of HPV genotypes in Polish couples, in which each woman is HPV positive, indicating a common infection of HPV 42, regardless of sex and anatomical site. These findings shed new light on HPV 42 significance, however they should be verified on a larger group of Polish participants, followed regularly in 6 months intervals, in oral as well as in genital areas.


Subject(s)
Mouth/virology , Oropharynx/virology , Papillomaviridae/genetics , Papillomavirus Infections/virology , Adult , Aged , Female , Foreskin/virology , Genotype , Humans , Male , Middle Aged , Papillomaviridae/isolation & purification , Papillomavirus Infections/epidemiology , Poland/epidemiology , Prevalence , Real-Time Polymerase Chain Reaction , Risk Factors , Sexual Partners , Sexually Transmitted Diseases/virology , Uterine Cervical Neoplasms/virology
9.
Mol Cell Proteomics ; 16(4 suppl 1): S92-S107, 2017 04.
Article in English | MEDLINE | ID: mdl-28179408

ABSTRACT

Herpes simplex virus (HSV-1) lytic infection results in global changes to the host cell proteome and the proteins associated with host chromatin. We present a system level characterization of proteome dynamics during infection by performing a multi-dimensional analysis during HSV-1 lytic infection of human foreskin fibroblast (HFF) cells. Our study includes identification and quantification of the host and viral proteomes, phosphoproteomes, chromatin bound proteomes and post-translational modifications (PTMs) on cellular histones during infection. We analyzed proteomes across six time points of virus infection (0, 3, 6, 9, 12 and 15 h post-infection) and clustered trends in abundance using fuzzy c-means. Globally, we accurately quantified more than 4000 proteins, 200 differently modified histone peptides and 9000 phosphorylation sites on cellular proteins. In addition, we identified 67 viral proteins and quantified 571 phosphorylation events (465 with high confidence site localization) on viral proteins, which is currently the most comprehensive map of HSV-1 phosphoproteome. We investigated chromatin bound proteins by proteomic analysis of the high-salt chromatin fraction and identified 510 proteins that were significantly different in abundance during infection. We found 53 histone marks significantly regulated during virus infection, including a steady increase of histone H3 acetylation (H3K9ac and H3K14ac). Our data provide a resource of unprecedented depth for human and viral proteome dynamics during infection. Collectively, our results indicate that the proteome composition of the chromatin of HFF cells is highly affected during HSV-1 infection, and that phosphorylation events are abundant on viral proteins. We propose that our epi-proteomics approach will prove to be important in the characterization of other model infectious systems that involve changes to chromatin composition.


Subject(s)
Chromatin/virology , Foreskin/virology , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Proteomics/methods , Viral Proteins/metabolism , Cells, Cultured , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Foreskin/cytology , Foreskin/metabolism , Fuzzy Logic , Gene Expression Regulation, Viral , HeLa Cells , Histones/metabolism , Humans , Male , Phosphorylation , Protein Processing, Post-Translational
10.
Zhonghua Nan Ke Xue ; 24(6): 516-519, 2018 Jun.
Article in Zh | MEDLINE | ID: mdl-30173456

ABSTRACT

OBJECTIVE: Human papilloma virus (HPV) is a necessary cause of cervical cancer and is also closely related to penile cancer, oropharyngeal cancer, and anal cancer in males. However, few studies are reported on male HPV. This study aimed to investigate HPV infection of the external genitalia in men whose female partners have cervical HPV infection. METHODS: We collected the relevant data on the male outpatients whose partners had cervical HPV infection in our Department of Urology and Andrology from August to December 2016. We obtained samples with nylon swabs from the glans penis, corona, inner layer of the prepuce and penile body and detected different types of HPV infection using the Hybribio HPV typing kit, PCR and membrane hybridization. RESULTS: Valid data were collected from 140 males, which showed 83.5% of HPV infection of the external genitalia, including 60 cases of HPV6 (43.2%), 27 cases of HPV16 (19.4%), 14 cases of HPV39 (10.1%), 13 cases of HPV18 (9.4%), 13 cases of HPV58 (9.4%), and 13 cases of HPV52 (9.4%). Redundant prepuce was found in 75.5% of the males, but there was no statistically significant difference in the incidence rate of HPV infection between the normal and redundant prepuce groups (P > 0.05). CONCLUSIONS: Men who have the female partners with positive cervical HPV are at high risk of HPV infection and therefore need to be screened and treated so as to reduce HPV infection in both sexes.


Subject(s)
Genital Diseases, Female/virology , Genital Diseases, Male/virology , Papillomavirus Infections/diagnosis , Penis/virology , Female , Foreskin/virology , Human papillomavirus 16/isolation & purification , Humans , Male , Papillomaviridae/isolation & purification , Penile Neoplasms/virology , Penis/abnormalities , Phimosis/virology , Polymerase Chain Reaction , Sexual Partners , Specimen Handling , Uterine Cervical Neoplasms/virology
11.
J Virol ; 90(12): 5824-5829, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27076640

ABSTRACT

UNLABELLED: The interferon (IFN)-mediated antiviral response is a central aspect of host defense; however, viruses have evolved multiple strategies to counteract IFN-mediated responses in order to successfully infect the host. Herpes simplex virus 1 (HSV-1), a typical human-restricted DNA virus, is capable of counteracting host immune responses via several distinct viral proteins, thus establishing a lifelong latent infection. In this study, we demonstrate that the VP24 protein, a serine protease of HSV-1 essential for the formation and maturation of capsids, is a novel antagonist of the beta interferon (IFN-ß) pathway. Here, VP24 was shown for the first time to dampen interferon stimulatory DNA (ISD)-triggered IFN-ß production and inhibit IFN-ß promoter activation induced by cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) and by STING, respectively. Further study demonstrated that ectopic expression of VP24 selectively blocked IFN regulatory factor 3 (IRF3) but not NF-κB promoter activation. In addition, VP24 was demonstrated to downregulate ISD-induced phosphorylation and dimerization of IRF3 during HSV-1 infection with a VP24 stable knockdown human foreskin fibroblast cell line. The underlying molecular mechanism is that VP24 abrogates the interaction between TANK-binding kinase 1 (TBK1) and IRF3, hence impairing IRF3 activation. These results illustrate that VP24 is able to block the production of IFN-ß by inhibiting IRF3 activation, which may represent a critical adaptation to enable viral effective replication within the host. IMPORTANCE: This study demonstrated that HSV-1 protein VP24 could inhibit IFN-ß production and promoter activation triggered by ISD, cGAS and STING and by STING, respectively. VP24 selectively blocked IRF3 promoter activation and ISD-induced phosphorylation and dimerization of IRF3 without affecting the NF-κB promoter activation during viral infection. VP24 also inhibited IRF3 activation by impeding the interaction between TBK1 and IRF3 during viral infection. This study provides new insights into the immune evasion mediated by HSV-1 and identifies VP24 as a crucial effector for HSV-1 to evade the host DNA-sensing signal pathway.


Subject(s)
DNA/metabolism , Herpesvirus 1, Human/metabolism , Interferon Regulatory Factor-3/metabolism , Signal Transduction , Viral Proteins/metabolism , Cell Line , Fibroblasts/virology , Foreskin/cytology , Foreskin/virology , Gene Knockdown Techniques , HEK293 Cells , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions , Humans , Immune Evasion , Interferon Regulatory Factor-3/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Male , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Transfection , Viral Proteins/genetics , Virus Replication
12.
PLoS Pathog ; 11(3): e1004729, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25748093

ABSTRACT

To gain insight into female-to-male HIV sexual transmission and how male circumcision protects against this mode of transmission, we visualized HIV-1 interactions with foreskin and penile tissues in ex vivo tissue culture and in vivo rhesus macaque models utilizing epifluorescent microscopy. 12 foreskin and 14 cadaveric penile specimens were cultured with R5-tropic photoactivatable (PA)-GFP HIV-1 for 4 or 24 hours. Tissue cryosections were immunofluorescently imaged for epithelial and immune cell markers. Images were analyzed for total virions, proportion of penetrators, depth of virion penetration, as well as immune cell counts and depths in the tissue. We visualized individual PA virions breaching penile epithelial surfaces in the explant and macaque model. Using kernel density estimated probabilities of localizing a virion or immune cell at certain tissue depths revealed that interactions between virions and cells were more likely to occur in the inner foreskin or glans penis (from local or cadaveric donors, respectively). Using statistical models to account for repeated measures and zero-inflated datasets, we found no difference in total virions visualized at 4 hours between inner and outer foreskins from local donors. At 24 hours, there were more virions in inner as compared to outer foreskin (0.0495 +/- 0.0154 and 0.0171 +/- 0.0038 virions/image, p = 0.001). In the cadaveric specimens, we observed more virions in inner foreskin (0.0507 +/- 0.0079 virions/image) than glans tissue (0.0167 +/- 0.0033 virions/image, p<0.001), but a greater proportion was seen penetrating uncircumcised glans tissue (0.0458 +/- 0.0188 vs. 0.0151 +/- 0.0100 virions/image, p = 0.099) and to significantly greater mean depths (29.162 +/- 3.908 vs. 12.466 +/- 2.985 µm). Our in vivo macaque model confirmed that virions can breach penile squamous epithelia in a living model. In summary, these results suggest that the inner foreskin and glans epithelia may be important sites for HIV transmission in uncircumcised men.


Subject(s)
Epithelial Cells/virology , Foreskin/virology , HIV Infections/immunology , HIV Infections/transmission , Animals , Cadaver , Circumcision, Male , Epithelial Cells/immunology , Female , Foreskin/immunology , HIV-1 , Humans , Immunohistochemistry , Macaca mulatta , Male , Penis/immunology , Penis/virology
13.
AIDS Res Ther ; 14(1): 49, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28893286

ABSTRACT

Circumcision reduces heterosexual HIV-1 acquisition in men by at least 60%. However, the biological mechanisms by which circumcision is protective remain incompletely understood. We test the hypothesis that the sub-preputial microenvironment created by the foreskin drives immune activation in adjacent foreskin tissues, facilitating HIV-1 infection through a combination of epithelial barrier disruption, enhanced dendritic cell maturation, and the recruitment/activation of neutrophils and susceptible CD4 T cell subsets such as Th17 cells. Furthermore, we provide evidence that the genital microbiome may be an important driver of this immune activation. This suggests that new modalities to reduce genital immune activation and/or alter the genital microbiome, used alone or in combination with topical microbicides, may be of significant benefit to HIV prevention.


Subject(s)
Circumcision, Male , Disease Susceptibility , HIV Infections/prevention & control , HIV-1/physiology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Chemokines/immunology , Foreskin/immunology , Foreskin/virology , HIV Infections/immunology , HIV Infections/virology , HIV-1/isolation & purification , Heterosexuality , Humans , Lymphocyte Activation , Male , Microbiota/immunology , Penis/cytology , Penis/immunology , Penis/virology , Primates , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Th17 Cells/immunology , Th17 Cells/virology
14.
Soc Stud Sci ; 47(5): 655-680, 2017 10.
Article in English | MEDLINE | ID: mdl-28639539

ABSTRACT

In this paper, I examine disputes over recent claims that male circumcision reduces HIV risk to suggest a complicated relationship between risk individualization and categorization. Whereas randomized controlled trials (RCTs) conducted in sub-Saharan Africa appear to have provided key evidence for the World Health Organization's endorsement of male circumcision as an HIV prevention strategy, RCTs alone did not provide evidence for the underlying causal mechanism. For that, medical authorities have turned to histo-immunological studies of the foreskin's biomolecular vulnerability to HIV, thus molecularizing risk. Some actors used these studies both as a way of shoring up results of RCTs conducted in sub-Saharan Africa and as an important rationale in arguments for making neonatal circumcision more widely available. Others, however, resisted this move to generalize the RCT results to other parts of the world, citing both contextual differences in HIV transmission patterns and conflicting scientific details regarding the biomolecular basis of the foreskin's susceptibility. Nevertheless, by locating an abstract notion of relative risk in the body itself, I argue that histological studies of foreskin have played a key role in stabilizing male circumcision status as a new risk category, largely independent of a given individual's risk profile.


Subject(s)
Circumcision, Male/ethics , Dissent and Disputes , Foreskin/surgery , HIV Infections/prevention & control , Adolescent , Adult , Africa South of the Sahara , Circumcision, Male/history , Dissent and Disputes/history , Female , Foreskin/virology , HIV Infections/history , HIV Infections/transmission , Health Policy/history , History, 20th Century , History, 21st Century , Humans , Male , Middle Aged , Randomized Controlled Trials as Topic , Risk Factors , Risk Management , Young Adult
15.
J Urol ; 195(6): 1784-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26796413

ABSTRACT

PURPOSE: There has been increasing interest in understanding the natural history of HPV and the diseases that it causes in men. HPV infection is strongly associated with penile cancer, lack of neonatal circumcision and phimosis. We investigated the incidence of HPV infection in asymptomatic men and patients with phimosis. MATERIALS AND METHODS: We assessed 110 asymptomatic men and 30 patients who underwent circumcision due to phimosis. DNA was extracted from swabbed samples collected from asymptomatic men and from foreskin samples collected at circumcision. Polymerase chain reaction using consensus primers for detecting HPV-MY09/11 was performed to detect generic HPV DNA. HPV genotyping was done by polymerase chain reaction amplification with primers for the E6 gene DNA sequences HPV6, HPV11, HPV16, HPV18, HPV31, HPV33, HPV35, HPV45 and HPV58. RESULTS: HPV was present in 46.66% of patients with phimosis, of whom 50% had high risk HPV genotypes. Of asymptomatic cases 16.36% were HPV positive but only 1 sample showed high risk HPV. We detected a significantly high rate of HPV genital infection in patients presenting with phimosis compared with asymptomatic men (p = 0.00167). The prevalence of high risk HPV genotypes in patients with phimosis was also statistically significant (p = 0.0004). CONCLUSIONS: We found a robust association between phimosis and the genital HPV prevalence in men and a significant frequency of high risk HPV. Other studies are needed to investigate the occurrence of factors that can increase the incidence of penile carcinoma and determine its impact on female genital infection in cervical cancer.


Subject(s)
Foreskin/virology , Papillomaviridae/genetics , Papillomavirus Infections/epidemiology , Phimosis/complications , Adolescent , Adult , Circumcision, Male , Cross-Sectional Studies , Foreskin/pathology , Genotype , Humans , Incidence , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Risk Factors , Young Adult
16.
Virol J ; 13: 137, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27496175

ABSTRACT

BACKGROUND: One of the most important functions of long noncoding RNAs (lncRNAs) is to control protein coding gene transcription by acting locally in cis, or remotely in trans. Herpes Simplex Virus type I (HSV-1) latently infects over 80 % of the population, its reactivation from latency usually results in productive infections in human epithelial cells, and is responsible for the common cold sores and genital Herpes. HSV-1 productive infection leads to profound changes in the host cells, including the host transcriptome. However, how genome wide lncRNAs expressions are affected by the infection and how lncRNAs expression relates to protein coding gene expression have not been analyzed. METHODS: We analyzed differentially expressed lncRNAs and their potential targets from RNA-seq data in HSV-1 infected human foreskin fibroblast (HFF) cells. Based on correlations of expression patterns of differentially expressed protein-coding genes and lncRNAs, we predicted that these lncRNAs may regulate, either in cis or in trans, the expression of many cellular protein-coding genes. RESULTS: Here we analyzed HSV-1 infection induced, differentially expressed lncRNAs and predicted their target genes. We detected 208 annotated and 206 novel differentially expressed lncRNAs. Gene Ontology and Pathway enrichment analyses revealed potential lncRNA targets, including genes in chromatin assembly, genes in neuronal development and neurodegenerative diseases and genes in the immune response, such as Toll-like receptor signaling and RIG-I-like receptor signaling pathways. CONCLUSIONS: We found that differentially expressed lncRNAs may regulate the expression of many cellular protein-coding genes involved in pathways from native immunity to neuronal development, thus revealing important roles of lncRNAs in the regulation of host transcriptional programs in HSV-1 infected human cells.


Subject(s)
Fibroblasts/metabolism , Foreskin/virology , Herpes Simplex/genetics , Herpesvirus 1, Human/physiology , RNA, Long Noncoding/genetics , Fibroblasts/virology , Foreskin/metabolism , Gene Expression Regulation , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Male , RNA, Long Noncoding/metabolism
17.
Arch Virol ; 161(5): 1273-84, 2016 May.
Article in English | MEDLINE | ID: mdl-26898402

ABSTRACT

Despite recent results of deletion experiments showing that open reading frame (ORF) UL49 of human cytomegalovirus (HCMV) is essential, the expression, function and functional location of its encoded protein remain unknown. We generated an antibody specific for pUL49 to investigate the protein product encoded by the UL49 ORF and identified its function in HCMV-infected host foreskin fibroblasts. A bacterial artificial chromosome (BAC) of HCMV strain Towne (pRV-Towne) and the UL49-deleted mutant pRV-delUL49Towne were used to observe virus growth by plaque assay. Using a UL49-protein-binding antibody, we located pUL49 in the fibroblast cytoplasm. pUL49 exhibited expression kinetics resembling those of the class ß-2 proteins and was detected in the virion tegument. Following deletion of UL49 ORF, the virus failed to replicate, but it could be recovered by addition of pUL49 from pCDNA3.1 (+)-UL49. Our findings indicate that UL49 ORF is essential for HCMV replication in host foreskin fibroblasts.


Subject(s)
Cytomegalovirus/physiology , Viral Proteins/physiology , Base Sequence , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus/growth & development , Cytomegalovirus Infections/virology , Fibroblasts/virology , Foreskin/cytology , Foreskin/virology , Gene Expression Regulation, Viral/physiology , Humans , Male , Microscopy, Fluorescence , Molecular Sequence Data , Viral Proteins/genetics , Virion/growth & development , Virion/physiology , Virus Replication/genetics , Virus Replication/physiology
18.
J Virol ; 88(3): 1714-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24257607

ABSTRACT

Epidemiological studies identified 12 high-risk HPV (hrHPV) types and 8 probable/possible hrHPV types that display different cancer risks. Functional studies on transforming properties of hrHPV are mainly limited to HPV16 and -18, which induce immortalization of human foreskin keratinocytes (HFKs) by successive bypass of two proliferative life span barriers, senescence and crisis. Here, we systematically compared the in vitro immortalization capacities, as well as influences on p53, pRb, hTERT, growth behavior, and differentiation capacity, of nine hrHPV types (HPV16, -18, -31, -33, -35, -45, -51, -52, and -59), and two probable hrHPV types (HPV66 and -70). By retroviral transduction, the respective E6/E7 coding sequences were expressed in HFKs from two or three independent donors. Reduced p53 levels and low-level hTERT expression in early-passage cells, as seen in HPV16-, -31-, -33-, and -35-, and to a lesser extent HPV18-transduced HFKs, was associated with continuous growth and an increased immortalization capacity. Less frequent immortalization by HPV45 and -51 and immortalization by HPV66 and -70 was preceded by an intervening period of strongly reduced growth (crisis) without prior increase in hTERT expression. Immortalization by HPV59 was also preceded by a period crisis, despite the onset of low hTERT expression at early passage. HPV52 triggered an extended life span but failed to induce immortality. Variations in p53 and pRb levels were not correlated with differences in alternative E6/E7 mRNA splicing in all hrHPV-transduced HFKs. On collagen rafts, transductants showed disturbed differentiation reminiscent of precancerous lesions. In conclusion, in vitro oncogenic capacities differ between the established hrHPV types, and both some established and probable hrHPV types display weak or moderate immortalization potential.


Subject(s)
Alphapapillomavirus/physiology , Cell Transformation, Viral , Papillomavirus Infections/virology , Alphapapillomavirus/genetics , Cells, Cultured , Foreskin/cytology , Foreskin/metabolism , Foreskin/virology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/virology , Male , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
J Virol ; 87(19): 10660-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23885077

ABSTRACT

Primary infection with human cytomegalovirus (HCMV) is generally asymptomatic in healthy individuals and results in a lifelong infection of the host. In contrast, in immunosuppressed transplant recipients and late-stage AIDS patients, HCMV infection and reactivation can result in severe disease or death. In vivo, latency is established in bone marrow CD34(+) progenitor cells with reactivation linked with their differentiation to macrophages and dendritic cells (DCs). However, previous analyses have relied on ex vivo differentiation of myeloid progenitor cells to DCs in culture. Here, we now report on the isolation and analysis of circulating blood myeloid DCs, resulting from natural differentiation in vivo, from healthy HCMV-seropositive carriers. We show that these in vivo-differentiated circulating DCs are fully permissive for HCMV and exhibit a phenotype similar to that of monocyte-derived DCs routinely used for in vitro studies of HCMV. Importantly, we also show that these DCs from healthy HCMV-seropositive donors carry HCMV genomes and, significantly, are typically positive for viral immediate-early (IE) gene expression, in contrast to circulating monocytes, which carry genomes with an absence of IE expression. Finally, we show that HCMV reactivation from these circulating DCs is enhanced by inflammatory stimuli. Overall, these data argue that the differentiation in vivo of myeloid progenitors to circulating DCs promotes the reactivation of HCMV lytic gene expression in healthy individuals, thereby providing valuable confirmation of studies performed using in vitro generation of DCs from myeloid precursors to study HCMV reactivation.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Dendritic Cells/virology , Virus Activation , Virus Latency , Cells, Cultured , Chromatin Immunoprecipitation , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , DNA, Viral/genetics , Dendritic Cells/metabolism , Dendritic Cells/pathology , Fibroblasts/cytology , Fibroblasts/virology , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Foreskin/cytology , Foreskin/virology , Humans , Male , Polymerase Chain Reaction , Seroepidemiologic Studies
20.
J Virol ; 87(19): 10489-500, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23885075

ABSTRACT

The human cytomegalovirus (HCMV) virion envelope contains a complex consisting of glycoproteins gH and gL plus proteins encoded by the UL128 locus (UL128L): pUL128, pUL130, and pUL131A. UL128L is necessary for efficient infection of myeloid, epithelial, and endothelial cells but limits replication in fibroblasts. Consequently, disrupting mutations in UL128L are rapidly selected when clinical isolates are cultured in fibroblasts. In contrast, bacterial artificial chromosome (BAC)-cloned strains TB40-BAC4, FIX, and TR do not contain overt disruptions in UL128L, yet no virus reconstituted from them has been reported to acquire mutations in UL128L in vitro. We performed BAC mutagenesis and reconstitution experiments to test the hypothesis that these strains contain subtle mutations in UL128L that were acquired during passage prior to BAC cloning. Compared to strain Merlin containing wild-type UL128L, all three strains produced higher yields of cell-free virus. Moreover, TB40-BAC4 and FIX spread cell to cell more rapidly than wild-type Merlin in fibroblasts but more slowly in epithelial cells. The differential growth properties of TB40-BAC4 and FIX (but not TR) were mapped to single-nucleotide substitutions in UL128L. The substitution in TB40-BAC4 reduced the splicing efficiency of UL128, and that in FIX resulted in an amino acid substitution in UL130. Introduction of these substitutions into Merlin dramatically increased yields of cell-free virus and increased cell-to-cell spread in fibroblasts but reduced the abundance of pUL128 in the virion and the efficiency of epithelial cell infection. These substitutions appear to represent mutations in UL128L that permit virus to be propagated in fibroblasts while retaining epithelial cell tropism.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Fibroblasts/virology , Membrane Glycoproteins/metabolism , Retinal Pigment Epithelium/virology , Viral Envelope Proteins/metabolism , Blotting, Western , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Endothelial Cells/metabolism , Endothelial Cells/virology , Fetus , Fibroblasts/metabolism , Foreskin/metabolism , Foreskin/virology , Genetic Variation , Humans , Male , Membrane Glycoproteins/genetics , Mutagenesis , Mutation/genetics , Plasmids/genetics , Retinal Pigment Epithelium/metabolism , Tropism , Viral Envelope Proteins/genetics , Virion/physiology , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL