Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Clin Infect Dis ; 78(2): 312-323, 2024 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-37738676

RESUMEN

BACKGROUND: The use of assays detecting cytomegalovirus (CMV)-specific T cell-mediated immunity may individualize the duration of antiviral prophylaxis after transplantation. METHODS: In this randomized trial, kidney and liver transplant recipients from 6 centers in Switzerland were enrolled if they were CMV-seronegative with seropositive donors or CMV-seropositive receiving antithymocyte globulins. Patients were randomized to a duration of antiviral prophylaxis based on immune monitoring (intervention) or a fixed duration (control). Patients in the control group were planned to receive 180 days (CMV-seronegative) or 90 days (CMV-seropositive) of valganciclovir. Patients were assessed monthly with a CMV ELISpot assay (T-Track CMV); prophylaxis in the intervention group was stopped if the assay was positive. The co-primary outcomes were the proportion of patients with clinically significant CMV infection and reduction in days of prophylaxis. Between-group differences were adjusted for CMV serostatus. RESULTS: Overall, 193 patients were randomized (92 in the immune-monitoring group and 101 in the control group), of whom 185 had evaluation of the primary outcome (87 and 98 patients). CMV infection occurred in 26 of 87 (adjusted percentage, 30.9%) in the immune-monitoring group and in 32 of 98 (adjusted percentage, 31.1%) in the control group (adjusted risk difference, -0.1; 95% confidence interval [CI], -13.0% to 12.7%; P = .064). The duration of prophylaxis was shorter in the immune-monitoring group (adjusted difference, -26.0 days; 95%, CI, -41.1 to -10.8 days; P < .001). CONCLUSIONS: Immune monitoring resulted in a significant reduction of antiviral prophylaxis, but we were unable to establish noninferiority of this approach on the co-primary outcome of CMV infection. CLINICAL TRIALS REGISTRATION: NCT02538172.


Asunto(s)
Infecciones por Citomegalovirus , Trasplante de Órganos , Humanos , Citomegalovirus , Antivirales/uso terapéutico , Monitorización Inmunológica , Infecciones por Citomegalovirus/diagnóstico , Receptores de Trasplantes , Trasplante de Órganos/efectos adversos , Ganciclovir/uso terapéutico
2.
Diagnostics (Basel) ; 13(18)2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37761277

RESUMEN

IVDR regulation represents a major challenge for diagnostic microbiology laboratories. IVDR complicates a broad range of aspects and poses a risk given the high diversity of pathogens (including rare but highly virulent microbes) and the large variety of samples submitted for analysis. The regular emergence of new pathogens (including Echovirus E-11, Adenovirus 41, Monkeypox virus, Alongshan virus, and Enterovirus D68, as recent examples in Europe in the post SARS-CoV-2 era) is another factor that makes IVDR regulation risky, because its detrimental effect on production of in-house tests will negatively impact knowledge and expertise in the development of new diagnostic tests. Moreover, such regulations negatively impact the availability of diagnostic tests, especially for neglected pathogens, and has a detrimental effect on the overall costs of the tests. The increased regulatory burden of IVDR may thereby pose an important risk for public health. Taken together, it will have a negative impact on the financial balance of diagnostic microbiology laboratories (especially small ones). The already-high standards of quality management of all ISO-accredited and Swissmedic-authorized laboratories render IVDR law of little value, at least in Switzerland, while tremendously increasing the regulatory burden and associated costs. Eventually, patients will need to pay for diagnostic assays outside of the framework of their insurance in order to obtain a proper diagnostic assessment, which may result in social inequity. Thus, based on the risk assessment outlined above, the coordinated commission for clinical microbiology proposes adjusting the IvDO ordinance by (i) introducing an obligation to be ISO 15189 accredited and (ii) not implementing the IvDO 2028 milestone.

4.
Ther Umsch ; 79(9): 448-453, 2022 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-36303534

RESUMEN

Lyme Disease - Laboratory Diagnostics Abstract. Lyme borreliosis is caused by Borrelia burgdorferi. Laboratory testing for Borrelia-specific antibodies is crucial for the diagnosis of Lyme borreliosis in addition to clinical definitions. The diagnostic approach consists of a two-tier testing: firstly, a highly sensitive screening test such as an enzyme immunoassay and secondly, a highly specific confirmatory assay such as a line immunoblot. The screening test detects Borrelia-specific IgM and IgG antibodies but also unspecific antibodies or cross-reactive antibodies against Treponema (causative agent of syphilis). Thus, a reactive screening test always needs confirmation by a specific test such as the immunoblot thereby resolving specific antibodies against different Borrelia antigens. Moreover, the characteristic spectrum of bands in the immunoblot provides evidence to divide the immune response into an early or a late stage of the disease. For the diagnosis of Lyme neuroborreliosis, intrathecal antibody production to B. burgdorferi should be determined by analyzing paired serum and cerebrospinal fluid samples obtained on the same timepoint. Diagnostics of Lyme borreliosis requires a comprehensive report of Borrelia-specific antibody responses and clinical manifestations.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme , Humanos , Anticuerpos Antibacterianos , Enfermedad de Lyme/diagnóstico , Antígenos Bacterianos , Inmunoglobulina G
5.
Plast Reconstr Surg Glob Open ; 10(4): e4230, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35415064

RESUMEN

Invasive fungal infections from Saksenaea, a fungus belonging to the Mucorales, have been rarely reported in central European climate zones. This study aims to raise awareness of invasive cutaneous infections with Saksenaea species. The first case of a cutaneous infection was diagnosed in Switzerland in an immunocompetent 79-year-old patient. A minor skin trauma of her left lower leg led to a fulminant infection causing necrosis and extensive loss of tissue. The combination of surgical debridement and administration of antifungal agents averted a prolonged course with a possible worse outcome. A pedicled hemisoleus muscle flap was used to reconstruct the defect and treatment was continued for 63 days. Methods: A systematic review in accordance with the Preferred Reporting Items for Systematic review and Meta-Analysis guidelines was conducted to identify all European cases of infection with Saksenaea species in immunocompetent hosts. The epidemiology, clinical presentation, microbiological diagnosis, and management of cases reported in Europe were summarized and analyzed. Conclusions: The prognosis of soft tissue infections with Saksenaea species. depends on early diagnosis and appropriate antifungal and surgical treatment. Reconstruction can be successful under ongoing antifungal treatment.

6.
Transfus Med Hemother ; 49(6): 338-345, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36654973

RESUMEN

Introduction: Screening of hepatitis B surface antigen (HBsAg) and individual-donation nucleic acid amplification testing (ID-NAT) of blood donors have become standard to detect hepatitis B virus (HBV) infection. However, there is still a residual risk of HBV transmission by blood components of donors suffering from occult HBV infection (OBI). Therefore, many countries implemented universal testing of anti-HBV core antigen (anti-HBc) antibodies in order to increase blood safety. In Switzerland, anti-HBc testing is not part of the routine blood donor-screening repertoire. Therefore, we sought to assess prevalence of donors with OBI in a Swiss blood donor collective. Methods: Blood donations were prospectively investigated for the presence of anti-HBc antibodies during two time periods (I: all donors, March 2017; II: first-time donors only, April 2017 until February 2018). Anti-HBc-positive findings were confirmed by an anti-HBc neutralization test. Discarded plasma samples of anti-HBc-confirmed positive donors were ultracentrifuged and subsequently retested by regular HBV-ID-NAT to search for traces of HBV. Results: During time period I, 78 (1.6%) individuals out of 4,923 donors were confirmed anti-HBc-positive. Sixty-nine (88%) anti-HBc-positive samples were available and processed by ultracentrifugation followed by repeat HBV-ID-NAT. Four samples (5.8%) were found positive for HBV DNA. Sixty-five (94.2%) samples remained HBV NAT-negative upon ultracentrifugation. During time period II, 56 (0.9%) donor samples out of 6,509 exhibited anti-HBc-confirmed positive. Fifty-five (98%) samples could be reassessed by HBV-ID-NAT upon ultracentrifugation. Three (5.5%) samples contained HBV DNA and 52 (94.5%) samples remained HBV NAT-negative. Conclusion: Overall, we detected 7 viremic OBI carriers among 11,432 blood donors, which tested negative for HBV by standard HBV-ID-NAT and HBsAg screening. In contrast, OBI carriers showed positive anti-HBc findings which could be confirmed in 83.8% of the cases. Thus, OBI might be missed by the current HBV screening process of Swiss blood donors. We suggest to review current HBV screening algorithm. Extended donor screening by anti-HBc testing may unmask OBI carriers and contribute to blood safety for the recipient of blood products.

7.
BMC Infect Dis ; 21(1): 1097, 2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34689748

RESUMEN

BACKGROUND: Aeromonas hydrophila is a gram-negative facultative anaerobic coccobacillus, which is an environmental opportunistic pathogen. A. hydrophila are involved in several infectious diseases such as gastroenteritis, septicemia and wound infections. However, gastroenteritis caused by Aeromonas spp. are rare and the clinical relevance of Aeromonas species in stool specimens is still under debate. CASE PRESENTATION: Our case concerns a 32-year-old woman who presented at hospital with a worsening watery diarrhea and fever requiring intensive care. A cholera-like illness was diagnosed. The patient had a past history of an anti-Hu syndrome with a myenteric ganglionitis. A molecular multiplex RT-PCR (QIAstat-Dx Gastrointestinal Panel, QIAGEN) covering a broad spectrum of diverse gastrointestinal pathogens performed directly from the stool was negative but the stool culture revealed growth of A. hydrophila. Further investigations of the A. hydrophila strain in cell cultures revealed the presence of a cytotoxic enterotoxin. CONCLUSIONS: Although A. hydrophila rarely causes gastroenteritis, Aeromonas spp. should be considered as a causative agent of severe gastroenteritis with a cholera-like presentation. This case highlights the need to perform culture methods from stool samples when PCR-based methods are negative and gastrointestinal infection is suspected.


Asunto(s)
Aeromonas , Gastroenteritis , Infecciones por Bacterias Gramnegativas , Adulto , Aeromonas hydrophila , Colectomía , Diarrea , Femenino , Gastroenteritis/diagnóstico , Infecciones por Bacterias Gramnegativas/diagnóstico , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Humanos
8.
Diagn Microbiol Infect Dis ; 101(1): 115424, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34111651

RESUMEN

Cartridge-based multiplex panels covering numerous pathogens offer an advantage of minimal hands-on-time and short time to result to commercial RT-PCR assays. In this study, we evaluated the performance of the ePlex respiratory pathogen panel (RPP) compared to the Fast Track Diagnostics Respiratory pathogens 21 multiplex RT-PCR assay (FTD21) using 400 clinical respiratory samples. Discrepant results were further analysed by a reference nucleic acid amplification testing (NAT) and a composite reference approach was used for final interpretation. Discordant results were observed in 56 targets corresponding to 54 samples. Sensitivities and specificities were 85.5% and 99.9% for the ePlex RPP and 95.8% and 99.7% for the FTD21 system, respectively. Altogether, the ePlex RPP is a valuable tool for the rapid detection of a number of different respiratory viruses with the exception of the coronavirus family (low sensitivity ranging from 50-80%) and samples with a low pathogen load (Ct values >33).


Asunto(s)
Técnicas de Diagnóstico Molecular , Reacción en Cadena de la Polimerasa Multiplex , Infecciones del Sistema Respiratorio/diagnóstico , Virus/aislamiento & purificación , Pruebas Diagnósticas de Rutina , Humanos , Infecciones del Sistema Respiratorio/virología , Sensibilidad y Especificidad , Factores de Tiempo , Virus/clasificación , Virus/genética
9.
Blood ; 137(23): 3225-3236, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33827115

RESUMEN

Primary immunodeficiencies in the costimulatory molecule CD27 and its ligand, CD70, predispose for pathologies of uncontrolled Epstein-Barr virus (EBV) infection in nearly all affected patients. We demonstrate that both depletion of CD27+ cells and antibody blocking of CD27 interaction with CD70 cause uncontrolled EBV infection in mice with reconstituted human immune system components. While overall CD8+ T-cell expansion and composition are unaltered after antibody blocking of CD27, only some EBV-specific CD8+ T-cell responses, exemplified by early lytic EBV antigen BMLF1-specific CD8+ T cells, are inhibited in their proliferation and killing of EBV-transformed B cells. This suggests that CD27 is not required for all CD8+ T-cell expansions and cytotoxicity but is required for a subset of CD8+ T-cell responses that protect us from EBV pathology.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Inmunidad Celular , Fosfoproteínas/inmunología , Transactivadores/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Linfocitos B/inmunología , Transformación Celular Viral/genética , Transformación Celular Viral/inmunología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Fosfoproteínas/genética , Transactivadores/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
10.
Eur J Immunol ; 51(1): 64-75, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32949466

RESUMEN

Immune responses to Epstein-Barr virus (EBV) infection synergize with the main genetic risk factor HLA-DRB1*15:01 (HLA-DR15) to increase the likelihood to develop the autoimmune disease multiple sclerosis (MS) at least sevenfold. In order to gain insights into this synergy, we investigated HLA-DR15 positive human immune compartments after reconstitution in immune-compromised mice (humanized mice) with and without EBV infection. We detected elevated activation of both CD4+ and CD8+ T cells in HLA-DR15 donor-reconstituted humanized mice at steady state, even when compared to immune compartments carrying HLA-DRB1*04:01 (HLA-DR4), which is associated with other autoimmune diseases. Increased CD8+ T cell expansion and activation was also observed in HLA-DR15 donor-reconstituted humanized mice after EBV infection. Despite this higher immune activation, EBV viral loads were less well controlled in the context of HLA-DR15. Indeed, HLA-DR15-restricted CD4+ T cell clones recognized EBV-transformed B cell lines less efficiently and demonstrated cross-reactivity toward allogeneic target cells and one MS autoantigen. These findings suggest that EBV as one of the main environmental risk factors and HLA-DR15 as the main genetic risk factor for MS synergize by priming hyperreactive T-cell compartments, which then control the viral infection less efficiently and contain cross-reactive CD4+ T cell clones.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Subtipos Serológicos HLA-DR/inmunología , Esclerosis Múltiple/inmunología , Inmunidad Adaptativa , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/virología , Predisposición Genética a la Enfermedad , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Subtipos Serológicos HLA-DR/genética , Herpesvirus Humano 4/inmunología , Humanos , Isoantígenos , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/genética , Vaina de Mielina/inmunología , Factores de Riesgo
11.
Life Sci Alliance ; 3(8)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32576602

RESUMEN

HIV and EBV are human pathogens that cause a considerable burden to worldwide health. In combination, these viruses are linked to AIDS-associated lymphomas. We found that EBV, which transforms B cells, renders them susceptible to HIV-1 infection in a CXCR4 and CD4-dependent manner in vitro and that CXCR4-tropic HIV-1 integrates into the genome of these B cells with the same molecular profile as in autologous CD4+ T cells. In addition, we established a humanized mouse model to investigate the in vivo interactions of EBV and HIV-1 upon coinfection. The respective mice that reconstitute human immune system components upon transplantation with CD34+ human hematopoietic progenitor cells could recapitulate aspects of EBV and HIV immunobiology observed in dual-infected patients. Upon coinfection of humanized mice, EBV/HIV dual-infected B cells could be detected, but were susceptible to CD8+ T-cell-mediated immune control.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/virología , Herpesvirus Humano 4/patogenicidad , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos B/virología , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Coinfección , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/virología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por VIH/genética , Seropositividad para VIH , VIH-1/metabolismo , VIH-1/patogenicidad , Células Madre Hematopoyéticas/patología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiología , Linfocitos T/inmunología
12.
Diagn Microbiol Infect Dis ; 96(2): 114924, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31757559

RESUMEN

Sapovirus (SaV) and astrovirus (AstV) increasingly are recognized as cause of acute viral gastroenteritis (AGE). We evaluated the real-time RT-PCR assays RIDA®GENE SaV and viral stool panel II (RGN RT-PCR) for detection of SaV, AstV, adenovirus (AdV) F40/41 and rotavirus (RoV) in clinical stool samples (n = 69). Results were compared with reference singleplex RT-PCRs. The sensitivity for SaV, AstV and RoV are 100%, the specificity ranges from 98.1% to 100%. In 10 out of 11 AdV (all types) samples, the RGN RT-PCR for AdV F40/41 displayed negative results. Retrospectively, 196 stool specimens from adult patients previously tested negative for norovirus (NoV) were analyzed. In about 10% of NoV-negative stool samples, AdV (n = 9), RoV (n = 6), AstV (n = 3) or SaV (n = 3) were found. The RGN RT-PCR assays are useful for detection of enteric viruses other than NoV. This study emphasizes the need for further testing of NoV-negative stool samples in patients with AGE.


Asunto(s)
Adenoviridae/genética , Heces/virología , Gastroenteritis/diagnóstico , Gastroenteritis/virología , Mamastrovirus/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Rotavirus/genética , Sapovirus/genética , Adenoviridae/clasificación , Adenoviridae/aislamiento & purificación , Adulto , Femenino , Humanos , Masculino , Mamastrovirus/clasificación , Mamastrovirus/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Reproducibilidad de los Resultados , Rotavirus/clasificación , Rotavirus/aislamiento & purificación , Sapovirus/clasificación , Sapovirus/aislamiento & purificación , Sensibilidad y Especificidad , Suiza
13.
Genes (Basel) ; 10(9)2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31470675

RESUMEN

Metagenomic next-generation sequencing (mNGS) can capture the full spectrum of viral pathogens in a specimen and has the potential to become an all-in-one solution for virus diagnostics. To date, clinical application is still in an early phase and limitations remain. Here, we evaluated the impact of viral mNGS for cases analyzed over two years in a tertiary diagnostics unit. High throughput mNGS was performed upon request by the treating clinician in cases where the etiology of infection remained unknown or the initial differential diagnosis was very broad. The results were compared to conventional routine testing regarding outcome and workload. In total, 163 specimens from 105 patients were sequenced. The main sample types were cerebrospinal fluid (34%), blood (33%) and throat swabs (10%). In the majority of the cases, viral encephalitis/meningitis or respiratory infection was suspected. In parallel, conventional virus diagnostic tests were performed (mean 18.5 individually probed targets/patients). mNGS detected viruses in 34 cases (32%). While often confirmatory, in multiple cases, the identified viruses were not included in the selected routine diagnostic tests. Two years of mNGS in a tertiary diagnostics unit demonstrated the advantages of a single, untargeted approach for comprehensive, rapid and efficient virus diagnostics, confirming the utility of mNGS in complementing current routine tests.


Asunto(s)
Metagenoma , Metagenómica/métodos , Técnicas de Diagnóstico Molecular/métodos , Análisis de Secuencia de ADN/métodos , Centros de Atención Terciaria/estadística & datos numéricos , Virosis/virología , Sangre/virología , Líquido Cefalorraquídeo/virología , Genoma Viral , Humanos , Metagenómica/estadística & datos numéricos , Técnicas de Diagnóstico Molecular/estadística & datos numéricos , Mucosa Bucal/virología , Análisis de Secuencia de ADN/estadística & datos numéricos , Virosis/diagnóstico , Virosis/epidemiología
15.
PLoS Pathog ; 15(5): e1007748, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31145756

RESUMEN

Epstein Barr virus (EBV) is one of the most ubiquitous human pathogens in the world, persistently infecting more than 90% of the adult human population. It drives some of the strongest human CD8+ T cell responses, which can be observed during symptomatic primary infection known as infectious mononucleosis (IM). Despite high viral loads and prolonged CD8+ T cell stimulation during IM, EBV enters latency and is under lifelong immune control in most individuals that experience this disease. We investigated whether changes in T cell function, as frequently characterized by PD-1 up-regulation, occur during IM due to the prolonged exposure to high antigen levels. We readily detected the expansion of PD-1 positive CD8+ T cells together with high frequencies of Tim-3, 2B4, and KLRG1 expression during IM and in mice with reconstituted human immune system components (huNSG mice) that had been infected with a high dose of EBV. These PD-1 positive CD8+ T cells, however, retained proliferation, cytokine production, and cytotoxic abilities. Multiple subsets of CD8+ T cells expanded during EBV infection, including PD-1+Tim-3+KLRG1+ cells that express CXCR5 and TCF-1 germinal center homing and memory markers, and may also contain BATF3. Moreover, blocking the PD-1 axis compromised EBV specific immune control and resulted in virus-associated lymphomagenesis. Finally, PD-1+, Tim-3+, and KLRG1+ CD8+ T cell expansion coincided with declining viral loads during low dose EBV infection. These findings suggest that EBV infection primes PD-1 positive CD8+ T cell populations that rely on this receptor axis for the efficient immune control of this ubiquitous human tumor virus.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Citotóxicos/inmunología , Carga Viral/inmunología , Adulto , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Estudios de Casos y Controles , Citocinas/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID
16.
Blood Adv ; 3(7): 1129-1144, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30952679

RESUMEN

Infectious mononucleosis, caused by infection with the human gamma-herpesvirus Epstein-Barr virus (EBV), manifests with one of the strongest CD8+ T-cell responses described in humans. The resulting T-cell memory response controls EBV infection asymptomatically in the vast majority of persistently infected individuals. Whether and how dendritic cells (DCs) contribute to the priming of this near-perfect immune control remains unclear. Here we show that of all the human DC subsets, plasmacytoid DCs (pDCs) play a central role in the detection of EBV infection in vitro and in mice with reconstituted human immune system components. pDCs respond to EBV by producing the interferon (IFN) subtypes α1, α2, α5, α7, α14, and α17. However, the virus curtails this type I IFN production with its latent EBV gene products EBNA3A and EBNA3C. The induced type I IFNs inhibit EBV entry and the proliferation of latently EBV-transformed B cells but do not influence lytic reactivation of the virus in vitro. In vivo, exogenous IFN-α14 and IFN-α17, as well as pDC expansion, delay EBV infection and the resulting CD8+ T-cell expansion, but pDC depletion does not significantly influence EBV infection. Thus, consistent with the observation that primary immunodeficiencies compromising type I IFN responses affect only alpha- and beta-herpesvirus infections, we found that EBV elicits pDC responses that transiently suppress viral replication and attenuate CD8+ T-cell expansion but are not required to control primary infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Interferón Tipo I/biosíntesis , Animales , Linfocitos T CD8-positivos/patología , Proliferación Celular , Humanos , Interferón Tipo I/farmacología , Ratones , Internalización del Virus/efectos de los fármacos , Replicación Viral
17.
Praxis (Bern 1994) ; 108(5): 335-339, 2019.
Artículo en Alemán | MEDLINE | ID: mdl-30940042

RESUMEN

Acute Viral Gastroenteritis: Viruses Other Than Norovirus Abstract. Norovirus is the leading cause of acute viral gastroenteritis. Norovirus is highly contagious, thus outbreaks of norovirus in hospitals and long-term care facilities are feared. Usually, stool samples of patients with a potentially viral gastroenteritis are first checked for the presence of norovirus. In recent years, sapovirus and astrovirus were increasingly reported as cause of acute gastroenteritis. Outbreaks of acute viral gastroenteritis caused by sapovirus or astrovirus are hardly distinguishable from those caused by norovirus because of a similar clinical presentation. Molecular analyses of stool specimen are needed for accurate diagnosis of the viral cause of acute gastroenteritis. It is worth to further investigate stool samples of patients suspected of acute viral gastroenteritis not only for norovirus, but also for sapovirus and astrovirus.


Asunto(s)
Infecciones por Astroviridae , Infecciones por Caliciviridae , Gastroenteritis , Sapovirus , Infecciones por Astroviridae/diagnóstico , Infecciones por Astroviridae/terapia , Infecciones por Caliciviridae/diagnóstico , Infecciones por Caliciviridae/terapia , Gastroenteritis/diagnóstico , Gastroenteritis/terapia , Gastroenteritis/virología , Humanos
18.
mBio ; 10(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30647153

RESUMEN

The human persistent and oncogenic Epstein-Barr virus (EBV) was one of the first viruses that were described to express viral microRNAs (miRNAs). These have been proposed to modulate many host and viral functions, but their predominant role in vivo has remained unclear. We compared recombinant EBVs expressing or lacking miRNAs during in vivo infection of mice with reconstituted human immune system components and found that miRNA-deficient EBV replicates to lower viral titers with decreased frequencies of proliferating EBV-infected B cells. In response, activated cytotoxic EBV-specific T cells expand to lower frequencies than during infection with miRNA-expressing EBV. However, when we depleted CD8+ T cells the miRNA-deficient virus reached similar viral loads as wild-type EBV, increasing by more than 200-fold in the spleens of infected animals. Furthermore, CD8+ T cell depletion resulted in lymphoma formation in the majority of animals after miRNA-deficient EBV infection, while no tumors emerged when CD8+ T cells were present. Thus, miRNAs mainly serve the purpose of immune evasion from T cells in vivo and could become a therapeutic target to render EBV-associated malignancies more immunogenic.IMPORTANCE Epstein-Barr virus (EBV) infects the majority of the human population and usually persists asymptomatically within its host. Nevertheless, EBV is the causative agent for infectious mononucleosis (IM) and for lymphoproliferative disorders, including Burkitt and Hodgkin lymphomas. The immune system of the infected host is thought to prevent tumor formation in healthy virus carriers. EBV was one of the first viruses described to express miRNAs, and many host and viral targets were identified for these in vitro However, their role during EBV infection in vivo remained unclear. This work is the first to describe that EBV miRNAs mainly increase viremia and virus-associated lymphomas through dampening antigen recognition by adaptive immune responses in mice with reconstituted immune responses. Currently, there is no prophylactic or therapeutic treatment to restrict IM or EBV-associated malignancies; thus, targeting EBV miRNAs could promote immune responses and limit EBV-associated pathologies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno , Evasión Inmune , MicroARNs/metabolismo , ARN Viral/metabolismo , Linfocitos T/inmunología , Animales , Linfocitos B/virología , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Eliminación de Gen , Humanos , Ratones , Ratones Transgénicos , MicroARNs/genética , ARN Viral/genética , Carga Viral
19.
Clin Infect Dis ; 69(6): 987-994, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30508036

RESUMEN

BACKGROUND: Before kidney transplantation, donors and recipients are routinely screened for viral pathogens using specific tests. Little is known about unrecognized viruses of the urinary tract that potentially result in transmission. Using an open metagenomic approach, we aimed to comprehensively assess virus transmission in living-donor kidney transplantation. METHODS: Living kidney donors and their corresponding recipients were enrolled at the time of transplantation. Follow-up study visits for recipients were scheduled 4-6 weeks and 1 year thereafter. At each visit, plasma and urine samples were collected and transplant recipients were evaluated for signs of infection or other transplant-related complications. For metagenomic analysis, samples were enriched for viruses, amplified by anchored random polymerase chain reaction (PCR), and sequenced using high-throughput metagenomic sequencing. Viruses detected by sequencing were confirmed using real-time PCR. RESULTS: We analyzed a total of 30 living kidney donor and recipient pairs, with a follow-up of at least 1 year. In addition to viruses commonly detected during routine post-transplant virus monitoring, metagenomic sequencing detected JC polyomavirus (JCPyV) in the urine of 7 donors and their corresponding recipients. Phylogenetic analysis confirmed infection with the donor strain in 6 cases, suggesting transmission from the transplant donor to the recipient, despite recipient seropositivity for JCPyV at the time of transplantation. CONCLUSIONS: Metagenomic sequencing identified frequent transmission of JCPyV from kidney transplant donors to recipients. Considering the high incidence rate, future studies within larger cohorts are needed to define the relevance of JCPyV infection and the donor's virome for transplant outcomes.


Asunto(s)
Virus JC/genética , Trasplante de Riñón/efectos adversos , Donadores Vivos , Metagenómica , Infecciones por Polyomavirus/epidemiología , Infecciones por Polyomavirus/etiología , Receptores de Trasplantes , Adulto , Comorbilidad , ADN Viral , Femenino , Alemania/epidemiología , Humanos , Inmunosupresores/efectos adversos , Virus JC/clasificación , Masculino , Metagenoma , Metagenómica/métodos , Persona de Mediana Edad , Infecciones por Polyomavirus/prevención & control , Infecciones por Polyomavirus/transmisión , Profilaxis Pre-Exposición , Prevalencia , Vigilancia en Salud Pública
20.
PLoS Pathog ; 14(4): e1007039, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29709016

RESUMEN

The oncogenic Epstein Barr virus (EBV) infects the majority of the human population and usually persists within its host for life without symptoms. The EBV oncoproteins nuclear antigen 3A (EBNA3A) and 3C (EBNA3C) are required for B cell transformation in vitro and are expressed in EBV associated immunoblastic lymphomas in vivo. In order to address the necessity of EBNA3A and EBNA3C for persistent EBV infection in vivo, we infected NOD-scid γcnull mice with reconstituted human immune system components (huNSG mice) with recombinant EBV mutants devoid of EBNA3A or EBNA3C expression. These EBV mutants established latent infection in secondary lymphoid organs of infected huNSG mice for at least 3 months, but did not cause tumor formation. Low level viral persistence in the absence of EBNA3A or EBNA3C seemed to be supported primarily by proliferation with the expression of early latent EBV gene products transitioning into absent viral protein expression without elevated lytic replication. In vitro, EBNA3A and EBNA3C deficient EBV infected B cells could be rescued from apoptosis through CD40 stimulation, mimicking T cell help in secondary lymphoid tissues. Thus, even in the absence of the oncogenes EBNA3A and 3C, EBV can access a latent gene expression pattern that is reminiscent of EBV persistence in healthy virus carriers without prior expression of its whole growth transforming program.


Asunto(s)
Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/patogenicidad , Animales , Linfocitos B/metabolismo , Células Cultivadas , Infecciones por Virus de Epstein-Barr/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA