Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biol Cell ; 114(7): 185-198, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35389514

RESUMO

MAGI-1 is a critical cellular scaffolding protein with over 110 different cellular and microbial protein interactors. Since the discovery of MAGI-1 in 1997, MAGI-1 has been implicated in diverse cellular functions such as polarity, cell-cell communication, neurological processes, kidney function, and a host of diseases including cancer and microbial infection. Additionally, MAGI-1 has undergone nomenclature changes in response to the discovery of an additional PDZ domain, leading to lack of continuity in the literature. We address the nomenclature of MAGI-1 as well as summarize many of the critical functions of the known interactions. Given the importance of many of the interactors, such as human papillomavirus E6, the Coxsackievirus and adenovirus receptor (CAR), and PTEN, the enhancement or disruption of MAGI-based interactions has the potential to affect cellular functions that can potentially be harnessed as a therapeutic strategy for a variety of diseases.


Assuntos
Domínios PDZ , Humanos
2.
J Virol ; 95(13): e0004621, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33762416

RESUMO

Adenoviruses (AdVs) are etiological agents of gastrointestinal, heart, eye, and respiratory tract infections that can be lethal for immunosuppressed people. Many AdVs use the coxsackievirus and adenovirus receptor (CAR) as a primary receptor. The CAR isoform resulting from alternative splicing that includes the eighth exon, CAREx8, localizes to the apical surface of polarized epithelial cells and is responsible for the initiation of AdV infection. We have shown that the membrane level of CAREx8 is tightly regulated by two MAGI-1 PDZ domains, PDZ2 and PDZ4, resulting in increased or decreased AdV transduction, respectively. We hypothesized that targeting the interactions between the MAGI-1 PDZ2 domain and CAREx8 would decrease the apical CAREx8 expression level and prevent AdV infection. Decoy peptides that target MAGI-1 PDZ2 were synthesized (TAT-E6 and TAT-NET1). PDZ2 binding peptides decreased CAREx8 expression and reduced AdV transduction. CAREx8 degradation was triggered by the activation of the regulated intramembrane proteolysis (RIP) pathway through a disintegrin and metalloproteinase (ADAM17) and γ-secretase. Further analysis revealed that ADAM17 interacts directly with the MAGI-1 PDZ3 domain, and blocking the PDZ2 domain enhanced the accessibility of ADAM17 to the substrate (CAREx8). Finally, we validated the efficacy of TAT-PDZ2 peptides in protecting the epithelia from AdV transduction in vivo using a novel transgenic animal model. Our data suggest that TAT-PDZ2 binding peptides are novel anti-AdV molecules that act by enhanced RIP of CAREx8 and decreased AdV entry. This strategy has additional translational potential for targeting other viral receptors that have PDZ binding domains, such as the angiotensin-converting enzyme 2 receptor. IMPORTANCE Adenovirus is a common threat in immunosuppressed populations and military recruits. There are no currently approved treatments/prophylactic agents that protect from most AdV infections. Here, we developed peptide-based small molecules that can suppress AdV infection of polarized epithelia by targeting the AdV receptor, coxsackievirus and adenovirus receptor (CAREx8). The newly discovered peptides target a specific PDZ domain of the CAREx8-interacting protein MAGI-1 and decrease AdV transduction in multiple polarized epithelial models. Peptide-induced CAREx8 degradation is triggered by extracellular domain (ECD) shedding through ADAM17 followed by γ-secretase-mediated nuclear translocation of the C-terminal domain. The enhanced shedding of the CAREx8 ECD further protected the epithelium from AdV infection. Taken together, these novel molecules protect the epithelium from AdV infection. This approach may be applicable to the development of novel antiviral molecules against other viruses that use a receptor with a PDZ binding domain.


Assuntos
Proteína ADAM17/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Adenoviridae/prevenção & controle , Moléculas de Adesão Celular/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/antagonistas & inibidores , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Guanilato Quinases/metabolismo , Células 3T3 , Adenoviridae/imunologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Cães , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Camundongos , Domínios Proteicos
3.
PLoS Pathog ; 16(4): e1008242, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32251490

RESUMO

Murine norovirus (MNoV) is an important model of human norovirus (HNoV) and mucosal virus infection more broadly. Viral receptor utilization is a major determinant of cell tropism, host range, and pathogenesis. The bona fide receptor for HNoV is unknown. Recently, we identified CD300lf as a proteinaceous receptor for MNoV. Interestingly, its paralogue CD300ld was also sufficient for MNoV infection in vitro. Here we explored whether CD300lf is the sole physiologic receptor in vivo and whether HNoV can use a CD300 ortholog as an entry receptor. We report that both CD300ld and CD300lf are sufficient for infection by diverse MNoV strains in vitro. We further demonstrate that CD300lf is essential for both oral and parenteral MNoV infection and to elicit anti-MNoV humoral responses in vivo. In mice deficient in STAT1 signaling, CD300lf is required for MNoV-induced lethality. Finally, we demonstrate that human CD300lf (huCD300lf) is not essential for HNoV infection, nor does huCD300lf inhibit binding of HNoV virus-like particles to glycans. Thus, we report huCD300lf is not a receptor for HNoV.


Assuntos
Infecções por Caliciviridae/virologia , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Norovirus/metabolismo , Receptores Imunológicos/metabolismo , Receptores Virais/metabolismo , Animais , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Norovirus/crescimento & desenvolvimento , Receptores Imunológicos/fisiologia , Tropismo Viral
4.
Biochem Cell Biol ; 99(1): 166-172, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32348689

RESUMO

Human noroviruses cause significant morbidity and mortality worldwide, but lack approved antivirals or vaccines to treat or prevent infections. The recent development of two cell culture systems in human transformed B cells (BJABs) and non-transformed human intestinal enteroid cultures overcomes a main limitation in identifying molecules with anti-norovirus activities. Lactoferrin is an iron-binding glycoprotein found in the milk of most mammals, with broad spectrum antimicrobial activities, including against the related murine norovirus in cell culture. In a Japanese clinical trial, ingestion of lactoferrin reduced the incidence of infectious gastroenteritis in the participants. Because human noroviruses were the most common cause of gastroenteritis in Japan during the clinical trial period, we sought to determine whether lactoferrin could inhibit infection with human norovirus. Our study, using a B cell culture model, demonstrates that lactoferrin reduces human norovirus infection. The mechanism of antiviral action is likely indirect and may involve the induction of innate interferon responses. Therefore, future studies are warranted to test the antiviral efficacy of lactoferrin against human norovirus infection in patients.


Assuntos
Antivirais/farmacologia , Lactoferrina/metabolismo , Norovirus/efeitos dos fármacos , Animais , Antivirais/química , Bovinos , Células Cultivadas , Humanos , Lactoferrina/química , Testes de Sensibilidade Microbiana , Replicação Viral/efeitos dos fármacos
5.
PLoS Pathog ; 15(10): e1008057, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31671153

RESUMO

Human astroviruses (HAstV) are understudied positive-strand RNA viruses that cause gastroenteritis mostly in children and the elderly. Three clades of astroviruses, classic, MLB-type and VA-type have been reported in humans. One limitation towards a better understanding of these viruses has been the lack of a physiologically relevant cell culture model that supports growth of all clades of HAstV. Herein, we demonstrate infection of HAstV strains belonging to all three clades in epithelium-only human intestinal enteroids (HIE) isolated from biopsy-derived intestinal crypts. A detailed investigation of infection of VA1, a member of the non-canonical HAstV-VA/HMO clade, showed robust replication in HIE derived from different patients and from different intestinal regions independent of the cellular differentiation status. Flow cytometry and immunofluorescence analysis revealed that VA1 infects several cell types, including intestinal progenitor cells and mature enterocytes, in HIE cultures. RNA profiling of VA1-infected HIE uncovered that the host response to infection is dominated by interferon (IFN)-mediated innate immune responses. A comparison of the antiviral host response in non-transformed HIE and transformed human colon carcinoma Caco-2 cells highlighted significant differences between these cells, including an increased magnitude of the response in HIE. Additional studies confirmed the sensitivity of VA1 to exogenous IFNs, and indicated that the endogenous IFN response of HIE to curtail the growth of strains from all three clades. Genotypic variation in the permissiveness of different HIE lines to HAstV could be overcome by pharmacologic inhibition of JAK/STAT signaling. Collectively, our data identify HIE as a universal infection model for HAstV and an improved model of the intestinal epithelium to investigate enteric virus-host interactions.


Assuntos
Infecções por Astroviridae/imunologia , Infecções por Astroviridae/veterinária , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Mamastrovirus/fisiologia , Tropismo Viral/genética , Animais , Células CACO-2 , Linhagem Celular , Chlorocebus aethiops , Enterócitos/virologia , Gastroenterite/virologia , Humanos , Imunidade Inata/imunologia , Interferons/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/virologia , Intestino Delgado/citologia , Intestino Delgado/virologia , Mamastrovirus/genética , Mamastrovirus/imunologia , Células Vero , Tropismo Viral/imunologia
6.
Mol Biol Evol ; 35(10): 2390-2400, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29955873

RESUMO

Viral evolutionary pathways are determined by the fitness landscape, which maps viral genotype to fitness. However, a quantitative description of the landscape and the evolutionary forces on it remain elusive. Here, we apply a biophysical fitness model based on capsid folding stability and antibody binding affinity to predict the evolutionary pathway of norovirus escaping a neutralizing antibody. The model is validated by experimental evolution in bulk culture and in a drop-based microfluidics that propagates millions of independent small viral subpopulations. We demonstrate that along the axis of binding affinity, selection for escape variants and drift due to random mutations have the same direction, an atypical case in evolution. However, along folding stability, selection and drift are opposing forces whose balance is tuned by viral population size. Our results demonstrate that predictable epistatic tradeoffs between molecular traits of viral proteins shape viral evolution.


Assuntos
Afinidade de Anticorpos , Evolução Biológica , Aptidão Genética , Modelos Genéticos , Norovirus/genética , Animais , Anticorpos Neutralizantes , Proteínas do Capsídeo/fisiologia , Epistasia Genética , Camundongos , Dobramento de Proteína , Estabilidade Proteica , Seleção Genética
8.
J Virol ; 90(3): 1499-506, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581993

RESUMO

UNLABELLED: A critical early step in murine norovirus (MNV) pathogenesis is crossing the intestinal epithelial barrier to reach the target cells for replication, i.e., macrophages, dendritic cells, and B cells. Our previous work showed that MNV replication decreases in the intestines of mice conditionally depleted of microfold (M) cells. To define the importance of Peyer's patch (PP) M cells during MNV pathogenesis, we used a model of BALB/c mice deficient in recombination-activating gene 2 (Rag2) and the common gamma chain (γc) (Rag-γc(-/-)), which lack gut-associated lymphoid tissues (GALT), such as Peyer's patches, and mature GP2(+) M cells. Rag-γc(-/-) mice were infected intraperitoneally or perorally with MNV-1 or CR3 for 24 or 72 h. Although the intestinal laminae propriae of Rag-γc(-/-) mice have a higher frequency of certain MNV target cells (dendritic cells and macrophages) than those of wild-type mice and lack others (B cells), Rag-γc(-/-) and wild-type BALB/c mice showed relatively similar viral loads in the intestine following infection by the intraperitoneal route, which provides direct access to target cells. However, Rag-γc(-/-) mice were not productively infected with MNV by the oral route, in which virions must cross the intestinal epithelial barrier. These data are consistent with a model whereby PP M cells are the primary route by which MNV crosses the intestinal epithelia of BALB/c mice. IMPORTANCE: Noroviruses (NoVs) are prevalent pathogens that infect their hosts via the intestine. Identifying key factors during the initial stages of virus infection in the host may provide novel points of intervention. Microfold (M) cells, antigen-sampling cells in the intestine, were previously shown to provide a gateway for murine NoV (MNV) into the host, but the relative importance of this uptake pathway remained unknown. Here we show that the absence of gut-associated lymphoid tissues (GALT), such as Peyer's patches, which contain high numbers of mature M cells, renders BALB/c mice refractory to oral infection with MNV. These findings are consistent with the model that M cells represent the primary route by which MNV crosses the intestinal epithelial barrier and infects underlying immune cells during a productive infection.


Assuntos
Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/virologia , Interações Hospedeiro-Patógeno , Norovirus/fisiologia , Nódulos Linfáticos Agregados/virologia , Internalização do Vírus , Animais , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C , Camundongos Knockout
10.
PLoS Pathog ; 11(3): e1004696, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25768646

RESUMO

Prevention of viral-induced respiratory disease begins with an understanding of the factors that increase or decrease susceptibility to viral infection. The primary receptor for most adenoviruses is the coxsackievirus and adenovirus receptor (CAR), a cell-cell adhesion protein normally localized at the basolateral surface of polarized epithelia and involved in neutrophil transepithelial migration. Recently, an alternate isoform of CAR, CAREx8, has been identified at the apical surface of polarized airway epithelia and is implicated in viral infection from the apical surface. We hypothesized that the endogenous role of CAREx8 may be to facilitate host innate immunity. We show that IL-8, a proinflammatory cytokine and a neutrophil chemoattractant, stimulates the protein expression and apical localization of CAREx8 via activation of AKT/S6K and inhibition of GSK3ß. Apical CAREx8 tethers infiltrating neutrophils at the apical surface of a polarized epithelium. Moreover, neutrophils present on the apical-epithelial surface enhance adenovirus entry into the epithelium. These findings suggest that adenovirus evolved to co-opt an innate immune response pathway that stimulates the expression of its primary receptor, apical CAREx8, to allow the initial infection the intact epithelium. In addition, CAREx8 is a new target for the development of novel therapeutics for both respiratory inflammatory disease and adenoviral infection.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae , Células Epiteliais/metabolismo , Imunidade Inata/imunologia , Infecções por Adenoviridae/metabolismo , Animais , Células Cultivadas , Epitélio/metabolismo , Humanos , Camundongos , Neutrófilos/imunologia , Receptores Virais/metabolismo
11.
J Virol ; 89(15): 7722-34, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25972549

RESUMO

UNLABELLED: Human noroviruses (HuNoVs) are positive-sense RNA viruses that can cause severe, highly infectious gastroenteritis. HuNoV outbreaks are frequently associated with recombination between circulating strains. Strain genotyping and phylogenetic analyses show that noroviruses often recombine in a highly conserved region near the junction of the viral polyprotein (open reading frame 1 [ORF1]) and capsid (ORF2) genes and occasionally within the RNA-dependent RNA polymerase (RdRP) gene. Although genotyping methods are useful for tracking changes in circulating viral populations, they report only the dominant recombinant strains and do not elucidate the frequency or range of recombination events. Furthermore, the relatively low frequency of recombination in RNA viruses has limited studies to cell culture or in vitro systems, which do not reflect the complexities and selective pressures present in an infected organism. Using two murine norovirus (MNV) strains to model coinfection, we developed a microfluidic platform to amplify, detect, and recover individual recombinants following in vitro and in vivo coinfection. One-step reverse transcriptase PCR (RT-PCR) was performed in picoliter drops with primers that identified the wild-type and recombinant progenies and scanned for recombination breakpoints at ∼1-kb intervals. We detected recombination between MNV strains at multiple loci spanning the viral protease, RdRP, and capsid ORFs and isolated individual recombinant RNA genomes that were present at a frequency of 1/300,000 or higher. This study is the first to examine norovirus recombination following coinfection of an animal and suggests that the exchange of RNA among viral genomes in an infected host occurs in multiple locations and is an important driver of genetic diversity. IMPORTANCE: RNA viruses increase diversity and escape host immune barriers by genomic recombination. Studies using a number of viral systems indicate that recombination occurs via template switching by the virus-encoded RNA-dependent RNA polymerase (RdRP). However, factors that govern the frequency and positions of recombination in an infected organism remain largely unknown. This work leverages advances in the applied physics of drop-based microfluidics to isolate and sequence rare recombinants arising from the coinfection of mice with two distinct strains of murine norovirus. This study is the first to detect and analyze norovirus recombination in an animal model.


Assuntos
Infecções por Caliciviridae/virologia , Norovirus/genética , Norovirus/isolamento & purificação , Recombinação Genética , Animais , Variação Genética , Genótipo , Humanos , Camundongos , Microfluídica , Dados de Sequência Molecular , Norovirus/classificação , Filogenia
12.
J Virol ; 88(8): 4543-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24501415

RESUMO

UNLABELLED: New human norovirus strains emerge every 2 to 3 years, partly due to mutations in the viral capsid that allow escape from antibody neutralization and herd immunity. To understand how noroviruses evolve antibody resistance, we investigated the structural basis for the escape of murine norovirus (MNV) from antibody neutralization. To identify specific residues in the MNV-1 protruding (P) domain of the capsid that play a role in escape from the neutralizing monoclonal antibody (MAb) A6.2, 22 recombinant MNVs were generated with amino acid substitutions in the A'B' and E'F' loops. Six mutations in the E'F' loop (V378F, A382K, A382P, A382R, D385G, and L386F) mediated escape from MAb A6.2 neutralization. To elucidate underlying structural mechanisms for these results, the atomic structure of the A6.2 Fab was determined and fitted into the previously generated pseudoatomic model of the A6.2 Fab/MNV-1 virion complex. Previously, two distinct conformations, A and B, of the atomic structures of the MNV-1 P domain were identified due to flexibility in the two P domain loops. A superior stereochemical fit of the A6.2 Fab to the A conformation of the MNV P domain was observed. Structural analysis of our observed escape mutants indicates changes toward the less-preferred B conformation of the P domain. The shift in the structural equilibrium of the P domain toward the conformation with poor structural complementarity to the antibody strongly supports a unique mechanism for antibody escape that occurs via antigen flexibility instead of direct antibody-antigen binding. IMPORTANCE: Human noroviruses cause the majority of all nonbacterial gastroenteritis worldwide. New epidemic strains arise in part by mutations in the viral capsid leading to escape from antibody neutralization. Herein, we identify a series of point mutations in a norovirus capsid that mediate escape from antibody neutralization and determine the structure of a neutralizing antibody. Fitting of the antibody structure into the virion/antibody complex identifies two conformations of the antibody binding domain of the viral capsid: one with a superior fit and the other with an inferior fit to the antibody. These data suggest a unique mode of antibody neutralization. In contrast to other viruses that largely escape antibody neutralization through direct disruption of the antibody-virus interface, we identify mutations that acted indirectly by limiting the conformation of the antibody binding loop in the viral capsid and drive the antibody binding domain into the conformation unable to be bound by the antibody.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Caliciviridae/imunologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Norovirus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Infecções por Caliciviridae/virologia , Proteínas do Capsídeo/genética , Linhagem Celular , Humanos , Evasão da Resposta Imune , Camundongos , Camundongos Knockout , Testes de Neutralização , Norovirus/química , Norovirus/genética
13.
Arch Virol ; 160(9): 2353-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26112762

RESUMO

Drug repurposing is a strategy employed to circumvent some of the bottlenecks involved in drug development, such as the cost and time needed for developing new molecular entities. Noroviruses cause recurrent epidemics and sporadic outbreaks of gastroenteritis associated with significant mortality and economic costs, but no treatment has been approved to date. Herein, a library of molecules previously used in humans was screened to find compounds with anti-noroviral activity. Antiviral testing for four selected compounds against murine norovirus infection revealed that rutin has anti-murine norovirus activity in cell-based assays.


Assuntos
Antivirais/farmacologia , Reposicionamento de Medicamentos , Norovirus/efeitos dos fármacos , Norovirus/fisiologia , Rutina/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Macrófagos/fisiologia , Macrófagos/virologia , Camundongos , Replicação Viral/efeitos dos fármacos
14.
J Gen Virol ; 95(Pt 9): 1958-1968, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24899153

RESUMO

Here, we report the isolation and functional characterization of mAbs against two murine norovirus (MNV) strains, MNV-1 and WU20, which were isolated following oral infection of mice. The mAbs were screened for reactivity against the respective homologous and heterologous MNV strain by ELISA. Selected mAbs were of IgA, IgG1, IgG2a or IgG2b isotype and showed a range of Western blot reactivities from non-binding to strong binding, suggesting recognition of conformational and linear epitopes. Some of the anti-MNV-1 antibodies neutralized both MNV-1 and WU20 infections in culture and in mice, but none of the anti-WU20 mAbs neutralized either virus. The non-neutralizing anti-MNV-1 IgG2b antibody 5C4.10 was mapped to the S domain of the MNV-1 capsid, whilst the epitopes of the neutralizing anti-MNV-1 IgA antibodies 2D3.7 and 4F9.4 were mapped to the P domain. Generation of neutralization escape viruses showed that two mutations (V339I and D348E) in the C'D' loop of the MNV-1 P domain mediated escape from mAb 2D3.7 and 4F9.4 neutralization. These findings broaden the known neutralizing epitopes of MNV to the main surface-exposed loops of the P domain. In addition, the current panel of antibodies provides valuable reagents for studying norovirus biology and development of diagnostic tools.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/imunologia , Norovirus/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Neutralizantes/genética , Especificidade de Anticorpos , Infecções por Caliciviridae/imunologia , Capsídeo/imunologia , Linhagem Celular , Epitopos/imunologia , Células HEK293 , Humanos , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Testes de Neutralização , Norovirus/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Carga Viral
15.
J West Afr Coll Surg ; 14(2): 229-232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38562392

RESUMO

Fibro-epithelial polyps (FEPs), also referred to as acrochordons or skin tags, are benign tumours that generally occur in women of reproductive age. They are uncommonly found in the vulva and vary in clinical appearances from small papillomatous growths to large pedunculated tumours. Typically, they are less than 5 cm. The wide range of morphological appearances of these tumours, especially when they are large, can be misinterpreted as malignant. This case involved a 30-year old multipara, 14-month post-partum who presented with a huge, irregular, firm, pedunculated mass on the right labium majus. The mass had patchy areas of skin ulceration and measured 25 cm × 15 cm × 10 cm on a 4-cm × 2-cm long stalk. It started as a 3-cm long finger-like projection with globular distal end that progressively increased in size over 7-month period. There were no swellings in other body parts. She had excisional biopsy of the mass which weighed 588 grams with histological diagnosis of inflamed FEP and had no recurrence at follow-up. This case illustrates an uncommon presentation of the second largest FEP of the vulva reported, which could be misinterpreted as malignant. Clinical, and pathological expertise with complete surgical excision are paramount for effective management to exclude atypia or malignancy and prevent recurrence.

16.
J West Afr Coll Surg ; 14(3): 355-357, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38988425

RESUMO

Acute abdomen due to incarcerated umbilical hernia is a surgical emergency. Acute abdomen secondary to gynaecological conditions is not uncommon. However, acute abdomen due to incarceration of a gynaecological tumour in an umbilical hernia is rare. A 25-year-old nullipara was admitted to the accident and emergency unit with a history of recurrent lower abdominal pain and abdominal swelling for 4 weeks. Pain worsened within the last 24 h necessitating presentation. Examination revealed a low-grade pyrexia, tachypnoea, an umbilical swelling with generalised abdominal tenderness, and a pelvic mass more in the right iliac fossa. Bedside abdomino-pelvic ultrasound scan confirmed bilateral adnexal masses with features suggestive of mature teratoma. A diagnosis of acute abdomen secondary to ovarian tumour accident was made. An emergency exploratory laparotomy revealed a huge right ovarian tumour incarcerated in an umbilical hernia. She had bilateral ovarian cystectomy and herniorrhaphy. Gynaecological tumours presenting as incarcerated or strangulated hernias are extremely rare but can be a cause of acute abdomen in women.

17.
J Virol ; 86(17): 9244-54, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718816

RESUMO

Epithelial integrity is essential for homeostasis and poses a formidable barrier to pathogen entry. Major factors for viral entry into epithelial cells are the localization and abundance of the primary receptor. The coxsackievirus and adenovirus receptor (CAR) is a primary receptor for these two pathogenic groups of viruses. In polarized epithelia, a low-abundance, alternatively spliced eight-exon isoform of CAR, CAR(Ex8), is localized apically where it can support viral infection from the air-exposed surface. Using biochemical, cell biology, genetic, and spectroscopic approaches, we show that the levels of apical CAR(Ex8) are negatively regulated by the PDZ domain-containing protein MAGI-1 (membrane-associated guanylate kinase with inverted orientation protein-1) and that two MAGI-1 PDZ domains, PDZ1 and PDZ3, regulate CAR(Ex8) levels in opposing ways. Similar to full-length MAGI-1, expression of the isolated PDZ3 domain significantly reduces cell surface CAR(Ex8) abundance and adenovirus infection. In contrast, the PDZ1 domain is able to rescue CAR(Ex8) and adenovirus infection from MAGI-1-mediated suppression. These data suggest a novel cell-based strategy to either suppress viral infection or augment adenovirus-based gene therapy.


Assuntos
Infecções por Adenoviridae/genética , Processamento Alternativo , Moléculas de Adesão Celular Neuronais/química , Moléculas de Adesão Celular Neuronais/metabolismo , Infecções por Coxsackievirus/genética , Éxons , Receptores Virais/genética , Proteínas Adaptadoras de Transdução de Sinal , Adenoviridae/fisiologia , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Animais , Moléculas de Adesão Celular , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Regulação para Baixo , Enterovirus/fisiologia , Guanilato Quinases , Humanos , Domínios PDZ , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/metabolismo
18.
J Gen Virol ; 93(Pt 1): 155-158, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21918008

RESUMO

Viruses are commonly investigated as vector systems for gene therapy. To be effective, virus-mediated gene-delivery systems require the presence of specific virus receptors to enter the target cell. One example is adenovirus and its primary receptor is the coxsackievirus and adenovirus receptor (CAR). Madin-Darby canine kidney (MDCK) cells have become a choice model system for studying CAR and adenovirus infection due to their ability to polarize rapidly into an epithelium with high transepithelial resistance. We show here that, whilst MDCK cells are resistant to adenovirus infection and hence appear functionally CAR-deficient, polarized MDCK cells express significant levels of CAR sequestered on the basolateral surface, where it is inaccessible for virus infection. Thus, although a cell type may be resistant to adenovirus infection, it is impossible to know whether it is due to a deficiency, as both CAR absence and inaccessibility are barriers to adenovirus-mediated gene transfer.


Assuntos
Infecções por Adenovirus Humanos/metabolismo , Adenovírus Humanos/genética , Vetores Genéticos/genética , Receptores Virais/metabolismo , Transdução Genética , Infecções por Adenovirus Humanos/genética , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/fisiologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Cães , Técnicas de Transferência de Genes , Vetores Genéticos/fisiologia , Humanos , Receptores Virais/genética
19.
Biochem Biophys Res Commun ; 425(1): 13-8, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22809504

RESUMO

We have previously shown that the Coxsackievirus and adenovirus receptor (CAR) can interact with post-synaptic density 95 (PSD-95) and localize PSD-95 to cell-cell junctions. We have also shown that activity of the acid sensing ion channel (ASIC3), a H(+)-gated cation channel that plays a role in mechanosensation and pain signaling, is negatively modulated by PSD-95 through a PDZ-based interaction. We asked whether CAR and ASIC3 simultaneously interact with PSD-95, and if so, whether co-expression of these proteins alters their cellular distribution and localization. Results indicate that CAR and ASIC3 co-immunoprecipitate only when co-expressed with PSD-95. CAR also brings both PSD-95 and ASIC3 to the junctions of heterologous cells. Moreover, CAR rescues PSD-95-mediated inhibition of ASIC3 currents. These data suggest that, in addition to activity as a viral receptor and adhesion molecule, CAR can play a role in trafficking proteins, including ion channels, in a PDZ-based scaffolding complex.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Canais Iônicos Sensíveis a Ácido/genética , Animais , Células COS , Chlorocebus aethiops , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Proteína 4 Homóloga a Disks-Large , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Domínios PDZ , Transporte Proteico
20.
Pathogens ; 11(2)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35215116

RESUMO

The coxsackievirus and adenovirus receptor (CAR) is an essential cellular protein that is involved in cell adhesion, cell signaling, and viral infection. The 8-exon encoded isoform (CAREx8) resides at the apical surface of polarized epithelia, where it is accessible as a receptor for adenovirus entering the airway lumen. Given its pivotal role in viral infection, it is a target for antiviral strategies. To understand the regulation of CAREx8 and determine the feasibility of receptor downregulation, the half-life of total and apical localized CAREx8 was determined and correlated with adenovirus transduction. Total and apical CAREx8 has a relatively short half-life of approximately 2 h. The half-life of apical CAREx8 correlates well with adenovirus transduction. These results suggest that antiviral strategies that aim to degrade the primary receptor for apical adenovirus infection will be effective within a relatively short time frame after application.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA