Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Allergy ; 77(1): 72-82, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33887070

RESUMEN

The ongoing COVID-19 pandemic caused by the SARS-CoV-2 coronavirus has affected the health of tens of millions of people worldwide. In particular, in elderly and frail individuals the infection can lead to severe disease and even fatal outcomes. Although the pandemic is primarily a human health crisis its consequences are much broader with a tremendous impact on global economics and social systems. Vaccines are considered the most powerful measure to fight the pandemic and protect people from COVID-19. Based on the concerted activities of scientists, manufacturers and regulators, the urgent need for effective countermeasures has provoked the development and licensure of novel COVID-19 vaccines in an unprecedentedly fast and flexible manner within <1 year. To ensure the safety and efficacy of these novel vaccines during the clinical development and the routine use in post-licensure vaccination campaigns existing regulatory requirements and procedures had to be wisely and carefully adapted to allow for an expedited evaluation without compromising the thoroughness of the regulatory and scientific assessment. In this review, we describe the regulatory procedures, concepts and requirements applied to guide and promote the highly accelerated development and licensure of safe and efficacious COVID-19 vaccines in Europe.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Anciano , Europa (Continente) , Humanos , Pandemias , SARS-CoV-2
2.
Artículo en Alemán | MEDLINE | ID: mdl-36264321

RESUMEN

The Paul-Ehrlich Institute (PEI) plays a central role in the release of vaccines in Germany as well as Europe. The experimental testing and release of each vaccine batch is carried out according to the procedures and regulations of the Official Control Authority Batch Release (OCABR) and the German medicine act paragraph 32. The independent testing aims to demonstrate the conformity of quality criteria set in the marketing authorization for each lot produced. This article illustrates both the batch release procedure in general and specifically for the newly developed and approved COVID-19 vaccines during the COVID-19 pandemic.


Asunto(s)
COVID-19 , Vacunas , Humanos , Alemania , Vacunas contra la COVID-19 , Pandemias/prevención & control , COVID-19/prevención & control , Vacunas/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 114(12): E2430-E2439, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28275093

RESUMEN

Hepatitis C virus (HCV) displays a restricted host species tropism and only humans and chimpanzees are susceptible to infection. A robust immunocompetent animal model is still lacking, hampering mechanistic analysis of virus pathogenesis, immune control, and prophylactic vaccine development. The closest homolog of HCV is the equine nonprimate hepacivirus (NPHV), which shares similar features with HCV and thus represents an animal model to study hepacivirus infections in their natural hosts. We aimed to dissect equine immune responses after experimental NPHV infection and conducted challenge experiments to investigate immune protection against secondary NPHV infections. Horses were i.v. injected with NPHV containing plasma. Flow cytometric analysis was used to monitor immune cell frequencies and activation status. All infected horses became viremic after 1 or 2 wk and viremia could be detected in two horses for several weeks followed by a delayed seroconversion and viral clearance. Histopathological examinations of liver biopsies revealed mild, periportally accentuated infiltrations of lymphocytes, macrophages, and plasma cells with some horses displaying subclinical signs of hepatitis. Following viral challenge, an activation of equine immune responses was observed. Importantly, after a primary NPHV infection, horses were protected against rechallenge with the homologous as well as a distinct isolate with only minute amounts of circulating virus being detectable.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/veterinaria , Enfermedades de los Caballos/inmunología , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Hepacivirus/clasificación , Hepacivirus/genética , Hepatitis C/inmunología , Hepatitis C/prevención & control , Hepatitis C/virología , Enfermedades de los Caballos/prevención & control , Enfermedades de los Caballos/virología , Caballos , Humanos , Filogenia , Linfocitos T/inmunología
4.
Artículo en Alemán | MEDLINE | ID: mdl-31776598

RESUMEN

Vaccinations are amongst the most important and powerful preventive measures modern medicine has to offer. By their nature, vaccines represent a very complex class of biological medicines. Licensure of novel vaccines is a process conducted on the basis of a comprehensive set of well-defined legal and procedural requirements. The key aim of the regulatory evaluation of vaccines is to confirm their pharmaceutical quality, safety, and efficacy in order to conclude on the positive benefit/risk ratio that is an absolute prerequisite for granting a license.In Europe there exist four types of licensing procedures for human vaccines (national, MRP, DCP, and centralized) depending on whether the vaccine is intended to be marketed nationally, in several, or all EU countries. Modern innovative vaccines are mostly licensed via the centralized EU procedure, which also offers a certain degree of procedural flexibility for specific vaccines under defined conditions. However, the basic regulatory requirements are the same for all types of licensing procedures. In order for a license to be granted, a vaccine has to fulfill all relevant regulatory requirements as regards pharmaceutical quality, including each manufacturing and control step as well as preclinical and clinical characterization. Most importantly, clinical trials in humans conducted prelicensure to determine vaccine safety and efficacy play a key role during the licensing procedure and for decision making.The WHO prequalification procedure was implemented to enable worldwide access to medicines of approved quality. Its prime aim is to establish and ensure appropriate universally recognized regulatory standards for vaccines to be used throughout the entire world.


Asunto(s)
Concesión de Licencias , Vacunación , Vacunas , Europa (Continente) , Alemania , Humanos , Organización Mundial de la Salud
5.
PLoS Pathog ; 12(7): e1005736, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27385030

RESUMEN

Hepatitis C virus (HCV) induces interferon (IFN) stimulated genes in the liver despite of distinct innate immune evasion mechanisms, suggesting that beyond HCV infected cells other cell types contribute to innate immune activation. Upon coculture with HCV replicating cells, human CD141+ myeloid dendritic cells (DC) produce type III IFN, whereas plasmacytoid dendritic cells (pDC) mount type I IFN responses. Due to limitations in the genetic manipulation of primary human DCs, we explored HCV mediated stimulation of murine DC subsets. Coculture of HCV RNA transfected human or murine hepatoma cells with murine bone marrow-derived DC cultures revealed that only Flt3-L DC cultures, but not GM-CSF DC cultures responded with IFN production. Cells transfected with full length or subgenomic viral RNA stimulated IFN release indicating that infectious virus particle formation is not essential in this process. Use of differentiated DC from mice with genetic lesions in innate immune signalling showed that IFN secretion by HCV-stimulated murine DC was independent of MyD88 and CARDIF, but dependent on TRIF and IFNAR signalling. Separating Flt3-L DC cultures into pDC and conventional CD11b-like and CD8α-like DC revealed that the CD8α-like DC, homologous to the human CD141+ DC, release interferon upon stimulation by HCV replicating cells. In contrast, the other cell types and in particular the pDC did not. Injection of human HCV subgenomic replicon cells into IFN-ß reporter mice confirmed the interferon induction upon HCV replication in vivo. These results indicate that HCV-replicating cells stimulate IFN secretion from murine CD8α-like DC independent of infectious virus production. Thus, this work defines basic principles of viral recognition by murine DC populations. Moreover, this model should be useful to explore the interaction between dendritic cells during HCV replication and to define how viral signatures are delivered to and recognized by immune cells to trigger IFN release.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Antígenos CD8/inmunología , Células Dendríticas/inmunología , Hepacivirus/inmunología , Interferón Tipo I/inmunología , Animales , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Hepatitis C/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
PLoS Pathog ; 12(4): e1005546, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27058035

RESUMEN

Human cytomegalovirus (HCMV) infections of healthy individuals are mostly unnoticed and result in viral latency. However, HCMV can also cause devastating disease, e.g., upon reactivation in immunocompromised patients. Yet, little is known about human immune cell sensing of DNA-encoded HCMV. Recent studies indicated that during viral infection the cyclic GMP/AMP synthase (cGAS) senses cytosolic DNA and catalyzes formation of the cyclic di-nucleotide cGAMP, which triggers stimulator of interferon genes (STING) and thus induces antiviral type I interferon (IFN-I) responses. We found that plasmacytoid dendritic cells (pDC) as well as monocyte-derived DC and macrophages constitutively expressed cGAS and STING. HCMV infection further induced cGAS, whereas STING expression was only moderately affected. Although pDC expressed particularly high levels of cGAS, and the cGAS/STING axis was functional down-stream of STING, as indicated by IFN-I induction upon synthetic cGAMP treatment, pDC were not susceptible to HCMV infection and mounted IFN-I responses in a TLR9-dependent manner. Conversely, HCMV infected monocyte-derived cells synthesized abundant cGAMP levels that preceded IFN-I production and that correlated with the extent of infection. CRISPR/Cas9- or siRNA-mediated cGAS ablation in monocytic THP-1 cells and primary monocyte-derived cells, respectively, impeded induction of IFN-I responses following HCMV infection. Thus, cGAS is a key sensor of HCMV for IFN-I induction in primary human monocyte-derived DC and macrophages.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Interferón Tipo I/biosíntesis , Monocitos/inmunología , Monocitos/virología , Nucleotidiltransferasas/inmunología , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Immunoblotting , Interferón Tipo I/inmunología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem , Transfección
7.
J Virol ; 89(6): 3200-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552725

RESUMEN

UNLABELLED: Worldwide, approximately 160 million people are chronically infected with hepatitis C virus (HCV), seven distinct genotypes of which are discriminated. The hallmarks of HCV are its genetic variability and the divergent courses of hepatitis C progression in patients. We assessed whether intragenotypic HCV variations would differentially trigger host innate immunity. To this end, we stimulated human primary plasmacytoid dendritic cells (pDC) with crude preparations of different cell culture-derived genotype 2a HCV variants. Parental Japanese fulminant hepatitis C virus (JFH1) did not induce interferon alpha (IFN-α), whereas the intragenotypic chimera Jc1 triggered massive IFN-α responses. Purified Jc1 retained full infectivity but no longer induced IFN-α. Coculture of pDC with HCV-infected hepatoma cells retrieved the capacity to induce IFN-α, whereas Jc1-infected cells triggered stronger responses than JFH1-infected cells. Since the infectivity of virus particles did not seem to affect pDC activation, we next tested Jc1 mutants that were arrested at different stages of particle assembly. These experiments revealed that efficient assembly and core protein envelopment were critically needed to trigger IFN-α. Of note, sequences within domain 2 of the core that vitally affect virus assembly also crucially influenced the IFN-α responses of pDC. These data showed that viral determinants shaped host innate IFN-α responses to HCV. IMPORTANCE: Although pegylated IFN-α plus ribavirin currently is the standard of care for the treatment of chronic hepatitis C virus infection, not much is known about the relevance of early interferon responses in the pathogenesis of hepatitis C virus infection. Here, we addressed whether intragenotypic variations of hepatitis C virus would account for differential induction of type I interferon responses mounted by primary blood-derived plasmacytoid dendritic cells. Surprisingly, a chimeric genotype 2a virus carrying the nonstructural genes of Japanese fulminant hepatitis C virus (JFH1) induced massive type I interferon responses, whereas the original genotype 2a JFH1 strain did not. Our detailed analyses revealed that, not the virus infectivity, but rather, the efficiency of virus assembly and core protein envelopment critically determined the magnitude of interferon responses. To our knowledge, this is the first example of hepatitis C virus-associated genetic variations that determine the magnitude of innate host responses.


Asunto(s)
Células Dendríticas/inmunología , Hepacivirus/fisiología , Hepatitis C/inmunología , Ensamble de Virus , Línea Celular , Células Dendríticas/virología , Femenino , Hepacivirus/química , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/virología , Humanos , Inmunidad Innata , Interferón-alfa , Masculino , Estructura Terciaria de Proteína , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología
8.
Hepatology ; 62(5): 1375-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26250868

RESUMEN

UNLABELLED: Patients carrying very rare loss-of-function mutations in interleukin-1 receptor-associated kinase 4 (IRAK4), a critical signaling mediator in Toll-like receptor signaling, are severely immunodeficient, highlighting the paramount role of IRAK kinases in innate immunity. We discovered a comparatively frequent coding variant of the enigmatic human IRAK2, L392V (rs3844283), which is found homozygously in ∼15% of Caucasians, to be associated with a reduced ability to induce interferon-alpha in primary human plasmacytoid dendritic cells in response to hepatitis C virus (HCV). Cytokine production in response to purified Toll-like receptor agonists was also impaired. Additionally, rs3844283 was epidemiologically associated with a chronic course of HCV infection in two independent HCV cohorts and emerged as an independent predictor of chronic HCV disease. Mechanistically, IRAK2 L392V showed intact binding to, but impaired ubiquitination of, tumor necrosis factor receptor-associated factor 6, a vital step in signal transduction. CONCLUSION: Our study highlights IRAK2 and its genetic variants as critical factors and potentially novel biomarkers for human antiviral innate immunity.


Asunto(s)
Hepatitis C Crónica/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Genotipo , Células HEK293 , Humanos , Interferón-alfa/biosíntesis , Interferones , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Interleucinas/genética , Polimorfismo de Nucleótido Simple , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/fisiología , Ubiquitinación
9.
J Immunol ; 193(5): 2496-503, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070849

RESUMEN

Upon treatment with vesicular stomatitis virus (VSV) particles, plasmacytoid dendritic cells (pDC) are triggered to mount substantial type I IFN responses, whereas myeloid DC (mDC) are only minor producers. Interestingly, bone marrow-derived (BM-)mDC were more vulnerable to infection with enhanced GFP (eGFP)-expressing VSV (VSVeGFP) than BM-pDC. BM-pDC stimulated with wild-type VSV mounted TLR-dependent IFN responses that were independent of RIG-I-like helicase (RLH) signaling. In contrast, in BM-pDC the VSV variant M2 induced particularly high IFN responses triggered in a TLR- and RLH-dependent manner, whereas BM-mDC stimulation was solely RLH-dependent. Importantly, VSVeGFP treatment of BM-pDC derived from IFN-ß yellow fluorescent protein (YFP) reporter mice (messenger of IFN-ß) resulted in YFP(+) and eGFP(+) single-positive cells, whereas among messenger of IFN-ß-BM-mDC most YFP(+) cells were also eGFP(+). This observation indicated that unlike mDC, direct virus infection was not required to trigger IFN responses of pDC. VSV-infected BM-mDC triggered BM-pDC to mount significantly higher IFN responses than free virus particles. Stimulation with infected cells enhanced the percentages of pDC subsets expressing either IFN-ß(+) or IFN-α6(+) plus IFN-ß(+). Irrespective of whether stimulated with free virus or infected cells, IFN induction was dependent on autophagy of pDC, whereas autophagy of the infected mDC was dispensable. Collectively, these results indicated that productive VSV infection was needed to trigger IFN responses of mDC, but not of pDC, and that IFN responses were primarily induced by virus-infected cells that stimulated pDC in a TLR-dependent manner.


Asunto(s)
Células Dendríticas/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Células Plasmáticas/inmunología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Células Dendríticas/patología , Interferón-alfa/genética , Interferón beta/genética , Ratones , Ratones Noqueados , Células Plasmáticas/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Estomatitis Vesicular/genética , Estomatitis Vesicular/patología
10.
J Virol ; 88(5): 2941-50, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371065

RESUMEN

UNLABELLED: The V proteins of paramyxoviruses control the innate immune response. In particular, the V protein of the genus Morbillivirus interferes with the signal transducer and activator of transcription 1 (STAT1), STAT2, and melanoma differentiation-associated protein 5 (mda5) signaling pathways. To characterize the contributions of these pathways to canine distemper virus (CDV) pathogenesis, we took advantage of the knowledge about the mechanisms of interaction between the measles virus V protein with these key regulators of innate immunity. We generated recombinant CDVs with V proteins unable to properly interact with STAT1, STAT2, or mda5. A virus with combined STAT2 and mda5 deficiencies was also generated, and available wild-type and V-protein-knockout viruses were used as controls. Ferrets infected with wild-type and STAT1-blind viruses developed severe leukopenia and loss of lymphocyte proliferation activity and succumbed to the disease within 14 days. In contrast, animals infected with viruses with STAT2 or mda5 defect or both STAT2 and mda5 defects developed a mild self-limiting disease similar to that associated with the V-knockout virus. This study demonstrates the importance of interference with STAT2 and mda5 signaling for CDV immune evasion and provides a starting point for the development of morbillivirus vectors with reduced immunosuppressive properties. IMPORTANCE: The V proteins of paramyxoviruses interfere with the recognition of the virus by the immune system of the host. For morbilliviruses, the V protein is known to interact with the signal transducer and activator of transcription 1 (STAT1) and STAT2 and the melanoma differentiation-associated protein 5 (mda5), which are involved in interferon signaling. Here, we examined the contribution of each of these signaling pathways to the pathogenesis of the carnivore morbillivirus canine distemper virus. Using viruses selectively unable to interfere with the respective signaling pathway to infect ferrets, we found that inhibition of STAT2 and mda5 signaling was critical for lethal disease. Our findings provide new insights in the mechanisms of morbillivirus immune evasion and may lead to the development of new vaccines and oncolytic vectors.


Asunto(s)
Virus del Moquillo Canino/patogenicidad , Moquillo/metabolismo , Interferones/metabolismo , ARN Helicasas/metabolismo , Factor de Transcripción STAT2/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Secuencia Conservada , Moquillo/inmunología , Virus del Moquillo Canino/genética , Virus del Moquillo Canino/inmunología , Hurones , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Unión Proteica , Transporte de Proteínas , Factor de Transcripción STAT1 , Transducción de Señal , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virulencia/genética , Replicación Viral
12.
Eur J Immunol ; 40(12): 3378-88, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108461

RESUMEN

Intestinal intraepithelial lymphocytes carrying the γδ TCR (γδ iIEL) are involved in the maintenance of epithelial integrity. γδ iIEL have an activated phenotype, characterized by CD69 expression and increased cell size compared with systemic T lymphocytes. As an additional activation marker, the majority of γδ iIEL express the CD8αα homodimer. However, our knowledge about cognate ligands for most γδ TCR remains fragmentary and recent advances show that γδ T cells including iIEL may be directly activated by cytokines or through NK-receptors, TLR and other pattern recognition receptors. We therefore asked whether the TCR of γδ iIEL was functional beyond its role during thymic selection. Using TcrdH2BeGFP (Tcrd, T-cell receptor δ locus; H2B, histone 2B) reporter mice to identify γδ T cells, we measured their intracellular free calcium concentration in response to TCR-crosslinking. In contrast to systemic γδ T cells, CD8αα(+) γδ iIEL showed high basal calcium levels and were refractory to TCR-dependent calcium-flux induction; however, they readily produced CC chemokine ligand 4 (CCL4) and IFN-γ upon TCR triggering in vitro. Notably, in vivo blocking of the γδ TCR with specific mAb led to a decrease of basal calcium levels in CD8αα(+) γδ iIEL. This suggests that the γδ TCR of CD8αα(+) γδ iIEL is constantly being triggered and therefore functional in vivo.


Asunto(s)
Quimiocina CCL4/metabolismo , Interferón gamma/metabolismo , Mucosa Intestinal/citología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos CD8/biosíntesis , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Células Cultivadas , Lectinas Tipo C/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T gamma-delta/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
13.
Vaccines (Basel) ; 9(7)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34358163

RESUMEN

Multiple preventive COVID-19 vaccines have been developed during the ongoing SARS coronavirus (CoV) 2 pandemic, utilizing a variety of technology platforms, which have different properties, advantages, and disadvantages. The acceleration in vaccine development required to combat the current pandemic is not at the expense of the necessary regulatory requirements, including robust and comprehensive data collection along with clinical product safety and efficacy evaluation. Due to the previous development of vaccine candidates against the related highly pathogenic coronaviruses SARS-CoV and MERS-CoV, the antigen that elicits immune protection is known: the surface spike protein of SARS-CoV-2 or specific domains encoded in that protein, e.g., the receptor binding domain. From a scientific point of view and in accordance with legal frameworks and regulatory practices, for the approval of a clinic trial, the Paul-Ehrlich-Institut requires preclinical testing of vaccine candidates, including general pharmacology and toxicology as well as immunogenicity. For COVID-19 vaccine candidates, based on existing platform technologies with a sufficiently broad data base, pharmacological-toxicological testing in the case of repeated administration, quantifying systemic distribution, and proof of vaccination protection in animal models can be carried out in parallel to phase 1 or 1/2 clinical trials. To reduce the theoretical risk of an increased respiratory illness through infection-enhancing antibodies or as a result of Th2 polarization and altered cytokine profiles of the immune response following vaccination, which are of specific concern for COVID-19 vaccines, appropriate investigative testing is imperative. In general, phase 1 (vaccine safety) and 2 (dose finding, vaccination schedule) clinical trials can be combined, and combined phase 2/3 trials are recommended to determine safety and efficacy. By applying these fundamental requirements not only for the approval and analysis of clinical trials but also for the regulatory evaluation during the assessment of marketing authorization applications, several efficacious and safe COVID-19 vaccines have been licensed in the EU by unprecedentedly fast and flexible procedures. Procedural and regulatory-scientific aspects of the COVID-19 licensing processes are described in this review.

14.
J Control Release ; 334: 201-212, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33865899

RESUMEN

Antibiotic treatment of tuberculosis (TB) is complex, lengthy, and can be associated with various adverse effects. As a result, patient compliance often is poor, thus further enhancing the risk of selecting multi-drug resistant bacteria. Macrophage mannose receptor (MMR)-positive alveolar macrophages (AM) constitute a niche in which Mycobacterium tuberculosis replicates and survives. Therefore, we encapsulated levofloxacin in lipid nanocarriers functionalized with fucosyl residues that interact with the MMR. Indeed, such nanocarriers preferentially targeted MMR-positive myeloid cells, and in particular, AM. Intracellularly, fucosylated lipid nanocarriers favorably delivered their payload into endosomal compartments, where mycobacteria reside. In an in vitro setting using infected human primary macrophages as well as dendritic cells, the encapsulated antibiotic cleared the pathogen more efficiently than free levofloxacin. In conclusion, our results point towards carbohydrate-functionalized nanocarriers as a promising tool for improving TB treatment by targeted delivery of antibiotics.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Antibacterianos/farmacología , Humanos , Lípidos , Macrófagos , Tuberculosis/tratamiento farmacológico
15.
Blood Adv ; 3(6): 839-850, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30867143

RESUMEN

Dendritic cells (DCs) take up antigen in the periphery, migrate to secondary lymphoid organs, and present processed antigen fragments to adaptive immune cells and thus prime antigen-specific immunity. During local inflammation, recirculating monocytes are recruited from blood to the inflamed tissue, where they differentiate to macrophages and DCs. In this study, we found that monocytes showed high transporter associated with antigen processing (TAP)-dependent peptide compartmentalization and that after antigen pulsing, they were not able to efficiently stimulate antigen-specific T lymphocytes. Nevertheless, upon in vitro differentiation to monocyte-derived DCs, TAP-dependent peptide compartmentalization as well as surface major histocompatibility complex I turnover decreased and the cells efficiently restimulated T lymphocytes. Although TAP-dependent peptide compartmentalization decreased during DC differentiation, TAP expression levels increased. Furthermore, TAP relocated from early endosomes in monocytes to the endoplasmic reticulum (ER) and lysosomal compartments in DCs. Collectively, these data are compatible with the model that during monocyte-to-DC differentiation, the subcellular relocation of TAP and the regulation of its activity assure spatiotemporal separation of local antigen uptake and processing by monocytes and efficient T-lymphocyte stimulation by DCs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Compartimento Celular , Diferenciación Celular/inmunología , Células Dendríticas/citología , Monocitos/citología , Presentación de Antígeno/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Lisosomas/metabolismo , Monocitos/inmunología , Linfocitos T/inmunología
16.
Cell Rep ; 25(1): 118-129.e4, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30282022

RESUMEN

In sterile neuroinflammation, a pathological role is proposed for microglia, whereas in viral encephalitis, their function is not entirely clear. Many viruses exploit the odorant system and enter the CNS via the olfactory bulb (OB). Upon intranasal vesicular stomatitis virus instillation, we show an accumulation of activated microglia and monocytes in the OB. Depletion of microglia during encephalitis results in enhanced virus spread and increased lethality. Activation, proliferation, and accumulation of microglia are regulated by type I IFN receptor signaling of neurons and astrocytes, but not of microglia. Morphological analysis of myeloid cells shows that type I IFN receptor signaling of neurons has a stronger impact on the activation of myeloid cells than of astrocytes. Thus, in the infected CNS, the cross talk among neurons, astrocytes, and microglia is critical for full microglia activation and protection from lethal encephalitis.


Asunto(s)
Astrocitos/inmunología , Encefalitis Viral/inmunología , Microglía/inmunología , Neuronas/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Astrocitos/patología , Comunicación Celular/inmunología , Encefalitis Viral/genética , Encefalitis Viral/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Neuronas/patología , Transducción de Señal
17.
Eur J Pharm Biopharm ; 95(Pt A): 13-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25701806

RESUMEN

Targeted drug delivery systems hold promise for selective provision of active compounds to distinct tissues or cell subsets. Thus, locally enhanced drug concentrations are obtained that would confer improved efficacy. As a consequence adverse effects should be diminished, as innocent bystander cells are less affected. Currently, several controlled drug delivery systems based on diverse materials are being developed. Some systems exhibit material-associated toxic effects and/or show low drug loading capacity. In contrast, liposomal nanocarriers are particularly favorable because they are well tolerated, poorly immunogenic, can be produced in defined sizes, and offer a reasonable payload capacity. Compared with other immune cells, professional antigen-presenting cells (APCs) demonstrate enhanced liposome uptake mediated by macropinocytosis, phagocytosis and presumably also by clathrin- and caveolae-mediated endocytosis. In order to further enhance the targeting efficacy toward APCs, receptor-mediated uptake appears advisable. Since APC subsets generally do not express single linage-specific receptors, members of the C-type lectin receptor (CLR) family are compelling targets. Examples of CLR expressed by APCs include DEC-205 (CD205) expressed by myeloid dendritic cells (DC) and monocytes, the mannose receptor C type 1 (MR, CD206) expressed by DC, monocytes and macrophages, DC-SIGN (CD209) expressed by DC, and several others. These receptors bind glycans, which are typically displayed by pathogens and thus support pathogen uptake and endocytosis. Further research will elucidate whether glycan-decorated liposomes will not only enhance APCs targeting but also enable preferential delivery of their payload to discrete subcellular compartments.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanosferas/metabolismo , Polisacáridos/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Liposomas , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Nanosferas/administración & dosificación , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Polisacáridos/administración & dosificación , Polisacáridos/inmunología
18.
Sci Rep ; 5: 18308, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26670584

RESUMEN

Treatment of PBMC with the CD4-specific mAb BT-061 induces CD4 down-modulation of T cells. Here we report that addition of BT-061 to purified T cells did not confer this effect, whereas incubation of T cells in BT-061 coated wells restored CD4 down-modulation. These results implied that Fcγ receptor mediated cell-cell interactions played a role. In consistence with this hypothesis PBMC depleted of CD64(+) monocytes did not confer CD4 down-modulation of BT-061 decorated T cells. Strikingly, CD4 down-modulation was observed in BT-061 treated synovial fluid punctuated from patients' inflamed joints that comprised enhanced numbers of CD64(+) cells. In contrast, in a circulating whole blood system injection of BT-061 did not induce CD4 down-modulation, due to CD64 saturation by serum IgG. Similarly, tonsil derived mononuclear cells devoid of CD64(+) cells did not show CD4 down-modulation, whereas addition of blood derived monocytes restored the effect. Thus, the interaction of BT-061 decorated T cells with CD64(+) cells is needed for CD4 down-modulation, implying that in patients BT-061 would primarily induce CD4 down-modulation at inflammatory sites. These results highlight the need not only to examine the interaction of a given mAb with single FcγR, but also the immunological environment that is appropriate to support such interactions.

19.
PLoS One ; 7(9): e45770, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049856

RESUMEN

Viruses of different families encode for regulators of the complement system (RCAs) or acquire such RCAs from the host to get protection against complement-mediated lysis (CML). As hepatitis C virus (HCV) shares no genetic similarity to any known RCA and is detectable at high titers in sera of infected individuals, we investigated whether HCV has adapted host-derived RCAs to resist CML. Here we report that HCV selectively incorporates CD59 while neither CD55, nor CD46 are associated with the virus. The presence of CD59 was shown by capture assays using patient- and cell culture-derived HCV isolates. Association of CD59 with HCV was further confirmed by Western blot analysis using purified viral supernatants from infected Huh 7.5 cells. HCV captured by antibodies specific for CD59 remained infectious for Huh 7.5 cells. In addition, blocking of CD59 in the presence of active complement reduced the titer of HCV most likely due to CML. HCV produced in CD59 knock-down cells were more significantly susceptible to CML compared to wild type virus, but neither replication, assembly nor infectivity of the virus seemed to be impaired in the absence of CD59. In summary our data indicate that HCV incorporates selectively CD59 in its envelope to gain resistance to CML in serum of infected individuals.


Asunto(s)
Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Hepacivirus/metabolismo , Proteína Cofactora de Membrana/metabolismo , Adulto , Anciano , Línea Celular , Línea Celular Tumoral , Activación de Complemento , Proteínas Inactivadoras de Complemento/inmunología , Proteínas del Sistema Complemento , Femenino , Citometría de Flujo/métodos , Genotipo , Humanos , Inmunoglobulina G/química , Masculino , Persona de Mediana Edad
20.
Mol Biotechnol ; 47(3): 262-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20938757

RESUMEN

Lentiviral gene transfer vectors are suitable for genetically modifying non-cycling primary human cells. In this study, we analyzed transduced human dendritic cells (DC) generated by the use of three different GFP-encoding lentiviral vectors, HIV-2 ROD A Δenv-GFP (ROD A), SIVsmm PBj ΔE EGFP (PBj), and SIVmac ΔE EGFP (SIVmac). CD14+ monocytes were isolated from buffy coat, transduced, and differentiated to immature and mature DC. Cytofluometric analysis of DC revealed high transduction efficiencies at MOI 1 for simian immunodeficiency virus (SIV)-derived vectors PBj and SIVmac ranging between 80-90 and 70-90%, respectively. In contrast, transduction with ROD A resulted only in approximately 30%-positive DC at the same MOI. Of note, none of the analyzed vectors affected expression of maturation and/or activation markers. Moreover, transduction with PBj or SIVmac did not induce significant cytokine responses whereas ROD A transduction stimulated weak interferon-alpha responses. SIVmac transduced DC showed normal phagocytosis of antigen and normal allo T cell stimulatory capacity when compared with untreated DC. Thus, the SIVmac lentiviral transduction vector is suitable for efficient genetic modification of human DC without affecting phenotype or function and thus qualifies this vector as a versatile tool for use in basic research.


Asunto(s)
Células Dendríticas/metabolismo , Vectores Genéticos/genética , Lentivirus/genética , Células Cultivadas , Citometría de Flujo , Humanos , Fagocitosis/genética , Fagocitosis/fisiología , Linfocitos T/citología , Linfocitos T/metabolismo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA