Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.520
Filter
1.
PLoS One ; 19(5): e0295088, 2024.
Article in English | MEDLINE | ID: mdl-38776332

ABSTRACT

Yaws affects children in tropical regions, while syphilis primarily affects sexually active adults worldwide. Despite various campaigns towards the eradication of yaws and elimination of syphilis, these two diseases are still present in Ghana. The aetiological agents of both diseases, two Treponema pallidum subspecies, are genetically similar. This study aimed to assess the prevalence of these treponematoses and the occurrence of pathogens causing similar skin lesions in the Ashanti region of Ghana. A point-of-care test was used to determine the seroprevalence of the treponematoses. Both yaws and syphilis were identified in the Ashanti region of Ghana. Multiplex PCR was used to identify treponemes and other pathogens that cause similar skin lesions. The results indicated that the seroprevalences of T. pallidum in individuals with yaws-like and syphilis-like lesions were 17.2% and 10.8%, respectively. Multiplex PCR results showed that 9.1%, 1.8% and 0.9% of yaws-like lesions were positive for Haemophilus ducreyi, herpes simplex virus-1 (HSV-1) and T. pallidum respectively. Among syphilis-like lesions, 28.3% were positive for herpes simplex virus -2 (HSV-2) by PCR. To our knowledge, this is the first time HSV-I and HSV-2 have been reported from yaws-like and syphilis-like lesions, respectively, in Ghana. The presence of other organisms apart from T. pallidum in yaws-like and syphilis-like lesions could impede the total healing of these lesions and the full recovery of patients. This may complicate efforts to achieve yaws eradication by 2030 and the elimination of syphilis and warrants updated empirical treatment guidelines for skin ulcer diseases.


Subject(s)
Haemophilus ducreyi , Syphilis , Treponema pallidum , Yaws , Humans , Ghana/epidemiology , Yaws/epidemiology , Yaws/microbiology , Syphilis/epidemiology , Syphilis/microbiology , Female , Adult , Male , Haemophilus ducreyi/isolation & purification , Haemophilus ducreyi/genetics , Adolescent , Prevalence , Treponema pallidum/genetics , Treponema pallidum/isolation & purification , Child , Young Adult , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/isolation & purification , Middle Aged , Seroepidemiologic Studies , Skin/microbiology , Skin/pathology , Skin/virology , Child, Preschool , Treponemal Infections/epidemiology , Treponemal Infections/microbiology
2.
Viruses ; 16(5)2024 04 26.
Article in English | MEDLINE | ID: mdl-38793562

ABSTRACT

The skin is a complex tissue that provides a strong physical barrier against invading pathogens. Despite this, many viruses can access the skin and successfully replicate in either the epidermal keratinocytes or dermal immune cells. In this review, we provide an overview of the antiviral T cell biology responding to cutaneous viral infections and how these responses differ depending on the cellular targets of infection. Much of our mechanistic understanding of T cell surveillance of cutaneous infection has been gained from murine models of poxvirus and herpesvirus infection. However, we also discuss other viral infections, including flaviviruses and papillomaviruses, in which the cutaneous T cell response has been less extensively studied. In addition to the mechanisms of successful T cell control of cutaneous viral infection, we highlight knowledge gaps and future directions with possible impact on human health.


Subject(s)
Skin Diseases, Viral , Skin , T-Lymphocytes , Humans , Animals , T-Lymphocytes/immunology , Skin Diseases, Viral/immunology , Skin Diseases, Viral/virology , Skin/virology , Skin/immunology , Mice , Immunologic Surveillance , Virus Diseases/immunology
3.
Viruses ; 16(5)2024 05 04.
Article in English | MEDLINE | ID: mdl-38793609

ABSTRACT

Dengue virus (DENV) is a continuing global threat that puts half of the world's population at risk for infection. This mosquito-transmitted virus is endemic in over 100 countries. When a mosquito takes a bloodmeal, virus is deposited into the epidermal and dermal layers of human skin, infecting a variety of permissive cells, including keratinocytes, Langerhans cells, macrophages, dermal dendritic cells, fibroblasts, and mast cells. In response to infection, the skin deploys an array of defense mechanisms to inhibit viral replication and prevent dissemination. Antimicrobial peptides, pattern recognition receptors, and cytokines induce a signaling cascade to increase transcription and translation of pro-inflammatory and antiviral genes. Paradoxically, this inflammatory environment recruits skin-resident mononuclear cells that become infected and migrate out of the skin, spreading virus throughout the host. The details of the viral-host interactions in the cutaneous microenvironment remain unclear, partly due to the limited body of research focusing on DENV in human skin. This review will summarize the functional role of human skin, the cutaneous innate immune response to DENV, the contribution of the arthropod vector, and the models used to study DENV interactions in the cutaneous environment.


Subject(s)
Dengue Virus , Dengue , Immunity, Innate , Skin , Animals , Humans , Cytokines/immunology , Cytokines/metabolism , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Dengue Virus/physiology , Host-Pathogen Interactions/immunology , Skin/virology , Skin/immunology , Virus Replication , Arthropods/virology
4.
Arch Dermatol Res ; 316(6): 292, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819524

ABSTRACT

Scientists from various areas of the world indicate in their studies that skin lesions occur in the course of infection with the SARS-CoV-2 virus. This article is a review of the most frequently described cutaneous manifestations of SARS-CoV-2 virus infection and the potential pathophysiology of their development, as well as information on abnormalities in histopathological tests. The article describes the impact of some factors related to the COVID-19 pandemic on the exacerbation of chronic dermatological diseases. This work was constructed on the basis of 142 research studies, reviews, and meta-analyses, focusing on the methods and materials used in individual works as well as the results and conclusions resulting from them. Some skin lesions may be a potential prognostic marker of the course of the disease and may also be a prodromal symptom or the only symptom of SARS-CoV-2 virus infection. Stress related to the COVID-19 pandemic may exacerbate some chronic dermatological diseases. A correlation was observed between the type of skin lesions and the patient's age. The occurrence of skin diseases may also be influenced by drugs used to treat infections caused by SARS-CoV-2. A relationship was observed between the patient's ethnic origin and skin lesions occurring in the course of COVID-19. There is a need to further diagnose the cutaneous manifestations of SARS-CoV-2 infection and to learn the detailed pathomechanism of their occurrence in order to better understand the essence of the disease and find an appropriate treatment method.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/complications , Adult , Young Adult , Skin Diseases/epidemiology , Skin Diseases/etiology , Skin/pathology , Skin/virology
5.
Vet Res ; 55(1): 54, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671518

ABSTRACT

This article reviews the avian viruses that infect the skin of domestic farm birds of primary economic importance: chicken, duck, turkey, and goose. Many avian viruses (e.g., poxviruses, herpesviruses, Influenza viruses, retroviruses) leading to pathologies infect the skin and the appendages of these birds. Some of these viruses (e.g., Marek's disease virus, avian influenza viruses) have had and/or still have a devasting impact on the poultry economy. The skin tropism of these viruses is key to the pathology and virus life cycle, in particular for virus entry, shedding, and/or transmission. In addition, for some emergent arboviruses, such as flaviviruses, the skin is often the entry gate of the virus after mosquito bites, whether or not the host develops symptoms (e.g., West Nile virus). Various avian skin models, from primary cells to three-dimensional models, are currently available to better understand virus-skin interactions (such as replication, pathogenesis, cell response, and co-infection). These models may be key to finding solutions to prevent or halt viral infection in poultry.


Subject(s)
Poultry Diseases , Virus Diseases , Animals , Poultry/virology , Poultry Diseases/virology , Skin/virology , Virus Diseases/veterinary , Virus Diseases/virology
6.
Front Immunol ; 15: 1366840, 2024.
Article in English | MEDLINE | ID: mdl-38680488

ABSTRACT

Rubella virus-associated granulomas commonly occur in immunocompromised individuals, exhibiting a diverse range of clinical presentations. These manifestations can vary from predominantly superficial cutaneous plaques or nonulcerative nodules to more severe deep ulcerative lesions, often accompanied by extensive necrosis and significant tissue destruction. TAP1 deficiency, an exceedingly rare primary immune-deficiency disorder, presents with severe chronic sino-pulmonary infection and cutaneous granulomas. This report highlights the occurrence of rubella virus-associated cutaneous granulomas in patients with TAP1 deficiency. Notably, the pathogenic mutation responsible for TAP1 deficiency stems from a novel genetic alteration that has not been previously reported. This novel observation holds potential significance for the field of diagnosis and investigative efforts in the context of immunodeficiency disorders.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 2 , Granuloma , Rubella virus , Humans , Granuloma/etiology , Granuloma/virology , Rubella virus/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Rubella/diagnosis , Rubella/immunology , Rubella/complications , Male , Mutation , Adult , Skin Diseases/etiology , Skin Diseases/virology , Female , Skin/pathology , Skin/virology
7.
Mamm Genome ; 35(2): 296-307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600211

ABSTRACT

Varicella-zoster virus (VZV), a common pathogen with humans as the sole host, causes primary infection and undergoes a latent period in sensory ganglia. The recurrence of VZV is often accompanied by severe neuralgia in skin tissue, which has a serious impact on the life of patients. During the acute infection of VZV, there are few related studies on the pathophysiological mechanism of skin tissue. In this study, transcriptome sequencing data from the acute response period within 2 days of VZV antigen stimulation of the skin were used to explore a model of the trajectory of skin tissue changes during VZV infection. It was found that early VZV antigen stimulation caused activation of mainly natural immune-related signaling pathways, while in the late phase activation of mainly active immune-related signaling pathways. JAK-STAT, NFκB, and TNFα signaling pathways are gradually activated with the progression of infection, while Hypoxia is progressively inhibited. In addition, we found that dendritic cell-mediated immune responses play a dominant role in the lesion damage caused by VZV antigen stimulation of the skin. This study provides a theoretical basis for the study of the molecular mechanisms of skin lesions during acute VZV infection.


Subject(s)
Herpesvirus 3, Human , Signal Transduction , Skin , Varicella Zoster Virus Infection , Herpesvirus 3, Human/genetics , Skin/pathology , Skin/virology , Skin/immunology , Animals , Varicella Zoster Virus Infection/virology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/genetics , Varicella Zoster Virus Infection/pathology , Humans , Mice , Dendritic Cells/immunology , Herpes Zoster/virology , Herpes Zoster/pathology , Herpes Zoster/genetics , Herpes Zoster/immunology , Transcriptome , Disease Models, Animal , Antigens, Viral/immunology , Antigens, Viral/genetics , NF-kappa B/metabolism , NF-kappa B/genetics
8.
Diagn Microbiol Infect Dis ; 109(2): 116268, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38513559

ABSTRACT

Mpox virus (MPXV) infection is difficult to distinguish from other (non-)infectious diseases. The etiology of rash can be differentiated by real-time polymerase chain reaction (rtPCR) on different types of samples. The study aims to provide experience with emerging MPXV diagnostics in a tertiary-level laboratory in Bosnia and Herzegovina. From July-December 2022, a total of 18 mpox suspected persons were tested. MPXV infection was confirmed by rtPCR in 10/18 (55.56 %) persons. The number of cases reached a peak in October 2022. The lowest median Crossing point (Cp) (x̄ = 29.76) was obtained from a swab of skin lesions in a viral transport medium (VTM). Evaluating the Cp values for the 7/9 mpox cases from whom paired swab samples from different anatomic sites were collected, higher positivity of skin lesion swabs in VTM was observed. In conclusion, our data highlighted the confirmatory role of rtPCR in the diagnosis of MPXV in skin lesion samples.


Subject(s)
DNA, Viral , Real-Time Polymerase Chain Reaction , Humans , Bosnia and Herzegovina , Real-Time Polymerase Chain Reaction/methods , DNA, Viral/genetics , DNA, Viral/isolation & purification , Male , Female , Adult , Mpox (monkeypox)/diagnosis , Mpox (monkeypox)/virology , Mpox (monkeypox)/epidemiology , Middle Aged , Adolescent , Young Adult , Tertiary Care Centers , Child , Monkeypox virus/genetics , Monkeypox virus/isolation & purification , Skin/virology , Molecular Diagnostic Techniques/methods
9.
J Virol ; 98(1): e0183023, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38088560

ABSTRACT

Usutu virus (USUV) and West Nile virus (WNV) are closely related emerging arboviruses belonging to the Flavivirus genus and posing global public health concerns. Although human infection by these viruses is mainly asymptomatic, both have been associated with neurological disorders such as encephalitis and meningoencephalitis. Since USUV and WNV are transmitted through the bite of an infected mosquito, the skin represents the initial site of virus inoculation and provides the first line of host defense. Although some data on the early stages of WNV skin infection are available, very little is known about USUV. Herein, USUV-skin resident cell interactions were characterized. Using primary human keratinocytes and fibroblasts, an early replication of USUV during the first 24 hours was shown in both skin cells. In human skin explants, a high viral tropism for keratinocytes was observed. USUV infection of these models induced type I and III interferon responses associated with upregulated expression of various interferon-stimulated genes as well as pro-inflammatory cytokine and chemokine genes. Among the four USUV lineages studied, the Europe 2 strain replicated more efficiently in skin cells and induced a higher innate immune response. In vivo, USUV and WNV disseminated quickly from the inoculation site to distal cutaneous tissues. In addition, viral replication and persistence in skin cells were associated with an antiviral response. Taken together, these results provide a better understanding of the pathophysiology of the early steps of USUV infection and suggest that the skin constitutes a major amplifying organ for USUV and WNV infection.IMPORTANCEUsutu virus (USUV) and West Nile virus (WNV) are closely related emerging Flaviviruses transmitted through the bite of an infected mosquito. Since they are directly inoculated within the upper skin layers, the interactions between the virus and skin cells are critical in the pathophysiology of USUV and WNV infection. Here, during the early steps of infection, we showed that USUV can efficiently infect two human resident skin cell types at the inoculation site: the epidermal keratinocytes and the dermal fibroblasts, leading to the induction of an antiviral innate immune response. Moreover, following cutaneous inoculation, we demonstrated that both viruses can rapidly spread, replicate, and persist in all distal cutaneous tissues in mice, a phenomenon associated with a generalized skin inflammatory response. These results highlight the key amplifying and immunological role of the skin during USUV and WNV infection.


Subject(s)
Flavivirus Infections , Flavivirus , Viral Tropism , West Nile Fever , West Nile virus , Animals , Humans , Mice , Antiviral Agents , Culicidae , Flavivirus Infections/virology , Interferons , West Nile Fever/virology , Skin/immunology , Skin/pathology , Skin/virology , In Vitro Techniques
10.
J Virol ; 97(12): e0127223, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38009914

ABSTRACT

IMPORTANCE: Human poxvirus infections have caused significant public health burdens both historically and recently during the unprecedented global Mpox virus outbreak. Although vaccinia virus (VACV) infection of mice is a commonly used model to explore the anti-poxvirus immune response, little is known about the metabolic changes that occur in vivo during infection. We hypothesized that the metabolome of VACV-infected skin would reflect the increased energetic requirements of both virus-infected cells and immune cells recruited to sites of infection. Therefore, we profiled whole VACV-infected skin using untargeted mass spectrometry to define the metabolome during infection, complementing these experiments with flow cytometry and transcriptomics. We identified specific metabolites, including nucleotides, itaconic acid, and glutamine, that were differentially expressed during VACV infection. Together, this study offers insight into both virus-specific and immune-mediated metabolic pathways that could contribute to the clearance of cutaneous poxvirus infection.


Subject(s)
Metabolic Reprogramming , Metabolome , Skin , Vaccinia virus , Vaccinia , Animals , Mice , Flow Cytometry , Gene Expression Profiling , Glutamine/metabolism , Mass Spectrometry , Nucleotides/metabolism , Skin/immunology , Skin/metabolism , Skin/virology , Vaccinia/immunology , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/metabolism , Viral Load
11.
Nat Commun ; 14(1): 3928, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402742

ABSTRACT

Tissue-resident memory (TRM) CD8+ T cells are largely derived from recently activated effector T cells, but the mechanisms that control the extent of TRM differentiation within tissue microenvironments remain unresolved. Here, using an IFNγ-YFP reporter system to identify CD8+ T cells executing antigen-dependent effector functions, we define the transcriptional consequences and functional mechanisms controlled by TCR-signaling strength that occur within the skin during viral infection to promote TRM differentiation. TCR-signaling both enhances CXCR6-mediated migration and suppresses migration toward sphingosine-1-phosphate, indicating the programming of a 'chemotactic switch' following secondary antigen encounter within non-lymphoid tissues. Blimp1 was identified as the critical target of TCR re-stimulation that is necessary to establish this chemotactic switch and for TRM differentiation to efficiently occur. Collectively, our findings show that access to antigen presentation and strength of TCR-signaling required for Blimp1 expression establishes the chemotactic properties of effector CD8+ T cells to promote residency within non-lymphoid tissues.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Receptors, Antigen, T-Cell , Skin , Virus Diseases , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Skin/immunology , Skin/virology , Virus Diseases/immunology , Cell Movement , Female , Animals , Mice , Mice, Inbred C57BL , Interferon-gamma/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Receptors, CXCR6/metabolism
12.
J Virol ; 96(17): e0099922, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000846

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Subject(s)
Alphavirus Infections , Arthritis , Myositis , Polysaccharides , Ross River virus , Skin , Alphavirus Infections/complications , Alphavirus Infections/immunology , Animals , Antiviral Agents/immunology , Arthritis/complications , Arthritis/immunology , Culicidae/virology , Dendritic Cells , Disease Models, Animal , Glycosylation , Humans , Mass Spectrometry , Mice , Monocytes , Myositis/complications , Myositis/immunology , Neutrophils , Polysaccharides/chemistry , Polysaccharides/immunology , Ross River virus/immunology , Skin/immunology , Skin/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
13.
J Virol ; 96(17): e0086422, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35969080

ABSTRACT

To infect its human host, herpes simplex virus 1 (HSV-1) must overcome the protective barriers of skin and mucosa. Here, we addressed whether pathological skin conditions can facilitate viral entry via the skin surface and used ex vivo infection studies to explore viral invasion in atopic dermatitis (AD) skin characterized by disturbed barrier functions. Our focus was on the visualization of the onset of infection in single cells to determine the primary entry portals in the epidermis. After ex vivo infection of lesional AD skin, we observed infected cells in suprabasal layers indicating successful invasion in the epidermis via the skin surface which was never detected in control skin where only sample edges allowed viral access. The redistribution of filaggrin, loricrin, and tight-junction components in the lesional skin samples suggested multiple defective mechanical barriers. To dissect the parameters that contribute to HSV-1 invasion, we induced an AD-like phenotype by adding the Th2 cytokines interleukin 4 (IL-4) and IL-13 to healthy human skin samples. Strikingly, we detected infected cells in the epidermis, implying that the IL-4/IL-13-driven inflammation is sufficient to induce modifications allowing HSV-1 to penetrate the skin surface. In summary, not only did lesional AD skin facilitate HSV-1 penetration but IL-4/IL-13 responses alone allowed virus invasion. Our results suggest that the defective epidermal barriers of AD skin and the inflammation-induced altered barriers in healthy skin can make receptors accessible for HSV-1. IMPORTANCE Herpes simplex virus 1 (HSV-1) can target skin to establish primary infection in the epithelium. While the human skin provides effective barriers against viral invasion under healthy conditions, a prominent example of successful invasion is the disseminated HSV-1 infection in the skin of atopic dermatitis (AD) patients. AD is characterized by impaired epidermal barrier functions, chronic inflammation, and dysbiosis of skin microbiota. We addressed the initial invasion process of HSV-1 in atopic dermatitis skin to understand whether the physical barrier functions are sufficiently disturbed to allow the virus to invade skin and reach its receptors on skin cells. Our results demonstrate that HSV-1 can indeed penetrate and initiate infection in atopic dermatitis skin. Since treatment of skin with IL-4 and IL-13 already resulted in successful invasion, we assume that inflammation-induced barrier defects play an important role for the facilitated access of HSV-1 to its target cells.


Subject(s)
Dermatitis, Atopic , Epidermis , Herpes Simplex , Herpesvirus 1, Human , Skin Diseases , Epidermis/pathology , Epidermis/virology , Herpes Simplex/pathology , Herpesvirus 1, Human/physiology , Humans , Inflammation , Interleukin-13 , Interleukin-4 , Skin/pathology , Skin/virology , Skin Diseases/virology , Tissue Culture Techniques
14.
J Virol ; 96(10): e0187521, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35475668

ABSTRACT

Persistent infection with some mucosal α-genus human papillomaviruses (HPVs; the most prevalent one being HPV16) can induce cervical carcinoma, anogenital cancers, and a subset of head and neck squamous cell carcinoma (HNSCC). Cutaneous ß-genus HPVs (such as HPV5 and HPV8) associate with skin lesions that can progress into squamous cell carcinoma with sun exposure in Epidermodysplasia verruciformis patients and immunosuppressed patients. Here, we analyzed mechanisms used by E6 proteins from the α- and ß-genus to inhibit the interferon-ß (IFNB1) response. HPV16 E6 mediates this effect by a strong direct interaction with interferon regulatory factor 3 (IRF3). The binding site of E6 was localized within a flexible linker between the DNA-binding domain and the IRF-activation domain of IRF3 containing an LxxLL motif. The crystallographic structure of the complex between HPV16 E6 and the LxxLL motif of IRF3 was solved and compared with the structure of HPV16 E6 interacting with the LxxLL motif of the ubiquitin ligase E6AP. In contrast, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3-binding domain (IBiD) of the CREB-binding protein (CBP), a key transcriptional coactivator in IRF3-mediated IFN-ß expression. IMPORTANCE Persistent HPV infections can be associated with the development of several cancers. The ability to persist depends on the ability of the virus to escape the host immune system. The type I interferon (IFN) system is the first-line antiviral defense strategy. HPVs carry early proteins that can block the activation of IFN-I. Among mucosal α-genus HPV types, the HPV16 E6 protein has a remarkable property to strongly interact with the transcription factor IRF3. Instead, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3 cofactor CBP. These results highlight the versatility of E6 proteins to interact with different cellular targets. The interaction between the HPV16 E6 protein and IRF3 might contribute to the higher prevalence of HPV16 than that of other high-risk mucosal HPV types in HPV-associated cancers.


Subject(s)
Interferon Regulatory Factor-3 , Interferon-beta , Oncogene Proteins, Viral , Papillomavirus Infections , Repressor Proteins , Human papillomavirus 16/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mucous Membrane/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Skin/virology
15.
Viruses ; 14(2)2022 02 18.
Article in English | MEDLINE | ID: mdl-35216020

ABSTRACT

To date, 14 human polyomaviruses (HPyVs) have been identified using high-throughput technologies. Among them, MCPyV, HPyV6, HPyV7 and TSPyV present a skin tropism, but a causal role in skin diseases has been established only for MCPyV as a causative agent of Merkel cell carcinoma (MCC) and TSPyV as an etiological agent of Trichodysplasia Spinulosa (TS). In the search for a possible role for cutaneous HPyVs in the development of skin malignant lesions, we investigated the prevalence of MCPyV, HPyV6, HPyV7 and TSPyV in actinic keratosis (AK), a premalignant skin lesion that has the potential to progress towards a squamous cell carcinoma (SCC). One skin lesion and one non-lesion skin from nine affected individuals were analyzed by qualitative PCR. MCPyV was detected in 9 out of 9 lesion biopsies and 6 out of 8 non-lesion biopsies. HPyV6 was detected only in healthy skin, while HPyV7 and TSPyV were not detected in any skin sample. These findings argue against a possible role of cutaneous HPyVs in AK. However, considering the small sample size analyzed, a definitive conclusion cannot be drawn. Longitudinal studies on large cohorts are warranted.


Subject(s)
Keratosis, Actinic/virology , Polyomavirus Infections/diagnosis , Polyomavirus/genetics , Skin/virology , Aged , Aged, 80 and over , Biopsy , DNA, Viral/genetics , High-Throughput Nucleotide Sequencing , Humans , Keratosis, Actinic/pathology , Male , Polyomavirus/classification , Polyomavirus/isolation & purification , Polyomavirus Infections/virology , Prevalence , Skin/pathology
16.
Int J Mol Sci ; 23(4)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35216085

ABSTRACT

The equine sarcoid is one of the most common neoplasias in the Equidae family. Despite the association of this tumor with the presence of bovine papillomavirus (BPV), the molecular mechanism of this lesion has not been fully understood. The transgenization of equine adult cutaneous fibroblast cells (ACFCs) was accomplished by nucleofection, followed by detection of molecular modifications using high-throughput NGS transcriptome sequencing. The results of the present study confirm that BPV-E4- and BPV-E1^E4-mediated nucleofection strategy significantly affected the transcriptomic alterations, leading to sarcoid-like neoplastic transformation of equine ACFCs. Furthermore, the results of the current investigation might contribute to the creation of in vitro biomedical models suitable for estimating the fates of molecular dedifferentiability and the epigenomic reprogrammability of BPV-E4 and BPV-E4^E1 transgenic equine ACFC-derived sarcoid-like cell nuclei in equine somatic cell-cloned embryos. Additionally, these in vitro models seem to be reliable for thoroughly recognizing molecular mechanisms that underlie not only oncogenic alterations in transcriptomic signatures, but also the etiopathogenesis of epidermal and dermal sarcoid-dependent neoplastic transformations in horses and other equids. For those reasons, the aforementioned transgenic models might be useful for devising clinical treatments in horses afflicted with sarcoid-related neoplasia of cutaneous and subcutaneous tissues.


Subject(s)
Fibroblasts/virology , Horse Diseases/virology , Horses/virology , Neoplasms/virology , Papillomaviridae/genetics , Sarcoidosis/virology , Skin Diseases/virology , Animals , Animals, Genetically Modified/virology , Equidae/virology , Papillomavirus Infections/virology , Skin/virology , Transcriptome/genetics
17.
Sci Rep ; 12(1): 1641, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102178

ABSTRACT

H84T BanLec is a molecularly engineered lectin cloned from bananas with broad-spectrum antiviral activity against several RNA viruses. H84T BanLec dimers bind glycoproteins containing high-mannose N-glycans on the virion envelope, blocking attachment, entry, uncoating, and spread. It was unknown whether H84T BanLec is effective against human herpesviruses varicella-zoster virus (VZV), human cytomegalovirus (HCMV), and herpes simplex virus 1 (HSV-1), which express high-mannose N-linked glycoproteins on their envelopes. We evaluated H84T BanLec against VZV-ORF57-Luc, TB40/E HCMV-fLuc-eGFP, and HSV-1 R8411 in cells, skin organ culture, and mice. The H84T BanLec EC50 was 0.025 µM for VZV (SI50 = 4000) in human foreskin fibroblasts (HFFs), 0.23 µM for HCMV (SI50 = 441) in HFFs, and 0.33 µM for HSV-1 (SI50 = 308) in Vero cells. Human skin was obtained from reduction mammoplasties and prepared for culture. Skin was infected and cultured up to 14 days. H84T BanLec prevented VZV, HCMV and HSV-1 spread in skin at 10 µM in the culture medium, and also exhibited dose-dependent antiviral effects. Additionally, H84T BanLec arrested virus spread when treatment was delayed. Histopathology of HCMV-infected skin showed no overt toxicity when H84T BanLec was present in the media. In athymic nude mice with human skin xenografts (NuSkin mice), H84T BanLec reduced VZV spread when administered subcutaneously prior to intraxenograft virus inoculation. This is the first demonstration of H84T BanLec effectiveness against DNA viruses. H84T BanLec may have additional unexplored activity against other, clinically relevant, glycosylated viruses.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Herpesviridae Infections/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 3, Human/drug effects , Plant Lectins/pharmacology , Skin Diseases, Viral/drug therapy , Skin/virology , Animals , Chlorocebus aethiops , Cytomegalovirus/growth & development , Herpesviridae Infections/virology , Herpesvirus 1, Human/growth & development , Herpesvirus 3, Human/growth & development , Mice, Nude , Musa/genetics , Plant Lectins/genetics , Skin Diseases, Viral/virology , Tissue Culture Techniques , Vero Cells , Virus Replication/drug effects
18.
J Allergy Clin Immunol ; 149(1): 388-399.e4, 2022 01.
Article in English | MEDLINE | ID: mdl-34033843

ABSTRACT

BACKGROUND: Rubella virus-induced granulomas have been described in patients with various inborn errors of immunity. Most defects impair T-cell immunity, suggesting a critical role of T cells in rubella elimination. However, the molecular mechanism of virus control remains elusive. OBJECTIVE: This study sought to understand the defective effector mechanism allowing rubella vaccine virus persistence in granulomas. METHODS: Starting from an index case with Griscelli syndrome type 2 and rubella skin granulomas, this study combined an international survey with a literature search to identify patients with cytotoxicity defects and granuloma. The investigators performed rubella virus immunohistochemistry and PCR and T-cell migration assays. RESULTS: This study identified 21 patients with various genetically confirmed cytotoxicity defects, who presented with skin and visceral granulomas. Rubella virus was demonstrated in all 12 accessible biopsies. Granuloma onset was typically before 2 years of age and lesions persisted from months to years. Granulomas were particularly frequent in MUNC13-4 and RAB27A deficiency, where 50% of patients at risk were affected. Although these proteins have also been implicated in lymphocyte migration, 3-dimensional migration assays revealed no evidence of impaired migration of patient T cells. Notably, patients showed no evidence of reduced control of concomitantly given measles, mumps, or varicella live-attenuated vaccine or severe infections with other viruses. CONCLUSIONS: This study identified lymphocyte cytotoxicity as a key effector mechanism for control of rubella vaccine virus, without evidence for its need in control of live measles, mumps, or varicella vaccines. Rubella vaccine-induced granulomas are a novel phenotype with incomplete penetrance of genetic disorders of cytotoxicity.


Subject(s)
Granuloma/etiology , Rubella Vaccine/adverse effects , T-Lymphocytes/immunology , Child , Child, Preschool , Female , Granuloma/genetics , Granuloma/immunology , Granuloma/virology , Humans , Infant , Phenotype , Rubella/genetics , Rubella/immunology , Rubella/virology , Skin/immunology , Skin/virology
19.
FEMS Microbiol Lett ; 368(20)2021 11 30.
Article in English | MEDLINE | ID: mdl-34849758

ABSTRACT

The prevalence of multidrug-resistant (MDR) strains has caused serious problems in the treatment of burn infections. MDR Enterobactercloacae and Enterobacterhormaechei have been defined as the causative agents of nosocomial infections in burn patients. In this situation, examination of phages side effects on human cell lines before any investigation on human or animal that can provide beneficial information about the safety of isolated phages. The aim of this study was to isolate and identify the specific bacteriophages on MDR E. cloacae and E. hormaechei isolated from burn wounds and to analyze the efficacy, cell viability and cell cytotoxicity of phages on A-375 and HFSF-PI cell lines by MTT (3-(4, 5-dimethylthiazol-2-yl)2,5-diphenyl-tetrazolium bromide) colorimetric assay and lactate dehydrogenase (LDH) release assay. Phages were isolated from urban sewage Isfahan, Iran. Enterobactercloacae strain Iau-EC100 (GenBank accession number: MZ314381) and E. hormaechei strain Iau-EHO100 (GenBank accession number: MZ348826) were sensitive to the isolated phages. Transmission electron microscopy (TEM) results revealed that PɸEn-CL and PɸEn-HO that were described had the morphologies of Myovirus and Inovirus, respectively. Overall, MTT and LDH assays showed moderate to excellent correlation in the evaluation of cytotoxicity of isolated phages. The results of MTT and LDH assays showed that, phages PɸEn-CL and PɸEn-HO had no significant toxicity effect on A375 and HFSF-PI 3 cells. Phage PɸEn-HO had a better efficacy on the two tested cell lines than other phage. Our results indicated that, there were significant differences between the two cytotoxicity assays in phage treatment compared to control.


Subject(s)
Bacteriophages , Burns , Enterobacter cloacae , Enterobacter , Wound Infection , Bacteriophages/physiology , Burns/complications , Burns/microbiology , Cell Line , Enterobacter/virology , Enterobacter cloacae/virology , Humans , Skin/microbiology , Skin/virology , Wound Infection/etiology , Wound Infection/microbiology
20.
PLoS One ; 16(12): e0261122, 2021.
Article in English | MEDLINE | ID: mdl-34914770

ABSTRACT

Fowlpox (FP) is an economically important viral disease of commercial poultry. The fowlpox virus (FPV) is primarily characterised by immunoblotting, restriction enzyme analysis in combination with PCR, and/or nucleotide sequencing of amplicons. Whole-genome sequencing (WGS) of FPV directly from clinical specimens prevents the risk of potential genome modifications associated with in vitro culturing of the virus. Only one study has sequenced FPV genomes directly from clinical samples using Nanopore sequencing, however, the study didn't compare the sequences against Illumina sequencing or laboratory propagated sequences. Here, the suitability of WGS for strain identification of FPV directly from cutaneous tissue was evaluated, using a combination of Illumina and Nanopore sequencing technologies. Sequencing results were compared with the sequence obtained from FPV grown in chorioallantoic membranes (CAMs) of chicken embryos. Complete genome sequence of FPV was obtained directly from affected comb tissue using a map to reference approach. FPV sequence from cutaneous tissue was highly similar to that of the virus grown in CAMs with a nucleotide identity of 99.8%. Detailed polymorphism analysis revealed the presence of a highly comparable number of single nucleotide polymorphisms (SNPs) in the two sequences when compared to the reference genome, providing essentially the same strain identification information. Comparative genome analysis of the map to reference consensus sequences from the two genomes revealed that this field isolate had the highest nucleotide identity of 99.5% with an FPV strain from the USA (Fowlpox virus isolate, FWPV-MN00.2, MH709124) and 98.8% identity with the Australian FPV vaccine strain (FWPV-S, MW142017). Sequencing results showed that WGS directly from cutaneous tissues is not only rapid and cost-effective but also provides essentially the same strain identification information as in-vitro grown virus, thus circumventing in vitro culturing.


Subject(s)
Chorioallantoic Membrane/virology , Fowlpox virus/isolation & purification , Fowlpox/diagnosis , Genome, Viral , High-Throughput Nucleotide Sequencing/methods , Skin/virology , Whole Genome Sequencing/methods , Animals , Australia , Chick Embryo , Chickens , Fowlpox/virology , Fowlpox virus/classification , Fowlpox virus/genetics , Fowlpox virus/growth & development , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...