Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 30(2): 745-762, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34450252

RESUMEN

The major challenge in the treatment of autoimmune diseases is the restoration of the impaired peripheral immune tolerance that always accompanies the development of such diseases. Here, we show that small splenic peptides (SSPs) of whole spleen extract efficiently suppress the development of psoriatic arthritis in vivo, even in the presence of sustained levels of pro-inflammatory cytokines. SSPs target dendritic cells (DCs) and convert them into tolerogenic cells, which in turn differentiate naive CD4+ cells into Foxp3-expressing T regulatory cells (Tregs). The latter requires direct contact between SSP-activated DCs and naive CD4+ T cells via PD-1 and CTLA4 immune checkpoint receptors of T cells. Finally, depletion of Foxp3+ Tregs in vivo abrogated the protective effect of SSPs on psoriatic arthritis development. We hypothesize that SSPs represent an intrinsic component of the adaptive immune system responsible for the physiological maintenance of peripheral tolerance and that therapeutically administered SSPs are able to restore imbalanced peripheral tolerance in autoimmune diseases.


Asunto(s)
Artritis Psoriásica , Tolerancia Inmunológica , Artritis Psoriásica/terapia , Citocinas , Células Dendríticas , Humanos , Tolerancia Periférica , Bazo , Linfocitos T Reguladores
2.
Oncoimmunology ; 10(1): 1885778, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33643696

RESUMEN

Non-small cell lung cancers (NSCLCs) establish a highly immunosuppressive tumor microenvironment supporting cancer growth. To interfere with cancer-mediated immunosuppression, selective immune-checkpoint inhibitors (ICIs) have been approved as a standard-of-care treatment for NSCLCs. However, the majority of patients poorly respond to ICI-based immunotherapies. Oncolytic viruses are amongst the many promising immunomodulatory treatments tested as standalone therapy or in combination with ICIs to improve therapeutic outcome. Previously, we demonstrated the oncolytic and immunomodulatory efficacy of low-pathogenic influenza Aviruses (IAVs) against NSCLCs in immunocompetent transgenic mice with alung-specific overexpression of active Raf kinase (Raf-BxB). IAV infection not only resulted in significant primary virus-induced oncolysis, but also caused afunctional reversion of tumor-associated macrophages (TAMs) comprising additional anti-cancer activity. Here we show that NSCLCs as well as TAMs and cytotoxic immune cells overexpress IC molecules of the PD-L2/PD-1 and B7-H3 signaling axes. Thus, we aimed to combine oncolytic IAV-infection with ICIs to exploit the benefits of both anti-cancer approaches. Strikingly, IAV infection combined with the novel B7-H3 ICI led to increased levels of M1-polarized alveolar macrophages and increased lung infiltration by cytotoxic Tlymphocytes, which finally resulted in significantly improved oncolysis of about 80% of existing tumors. In contrast, application of clinically approved α-PD-1 IC antibodies alone or in combination with oncolytic IAV did not provide additional oncolytic or immunomodulatory efficacy. Thus, individualized therapy with synergistically acting oncolytic IAV and B7-H3 ICI might be an innovative future approach to target NSCLCs that are resistant to approved ICIs in patients.


Asunto(s)
Gripe Humana , Neoplasias Pulmonares , Virus Oncolíticos , Orthomyxoviridae , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico , Pulmón , Neoplasias Pulmonares/terapia , Ratones , Virus Oncolíticos/genética , Microambiente Tumoral
3.
Am J Pathol ; 188(5): 1236-1245, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29458009

RESUMEN

Four-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein with fine-tuning adjustment properties. It acts as a regulator of signaling cascades but also as a cofactor of transcription and controls several anti-inflammatory immune responses. Recently, we described FHL2 as a novel regulator of influenza A virus propagation. We have shown that in vitro FHL2 restricts viral replication by accelerating the interferon regulatory factor 3-dependent transcription of the Ifnb1 gene. In this work, we unraveled an ambiguous role of FHL2 during influenza A virus infection in vivo. Although FHL2 restrained viral replication during the first 24 hours of infection, it significantly delayed viral clearance afterward. Comparison of lung immune status of wild-type and FHL2 knockout mice during influenza virus infection did not acknowledge significant differences in the innate host immune response but revealed an improved migration of dendritic cells from infected lungs into draining lymph nodes as well as increased levels of activated CD8+ T lymphocytes accumulated in the lungs of FHL2 knockout mice.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inflamación/metabolismo , Virus de la Influenza A , Proteínas con Homeodominio LIM/metabolismo , Pulmón/metabolismo , Proteínas Musculares/metabolismo , Factores de Transcripción/metabolismo , Animales , Movimiento Celular/fisiología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Inflamación/patología , Inflamación/virología , Proteínas con Homeodominio LIM/genética , Pulmón/patología , Pulmón/virología , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Factores de Transcripción/genética , Replicación Viral/fisiología
4.
Am J Pathol ; 187(11): 2388-2398, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28823868

RESUMEN

Four and a half LIM domain protein 2 (Fhl2) is an intracellular adaptor molecule with a high protein-protein interaction capacity. It acts as a modulator of several signaling molecules in the cytosol and as a cofactor of transcription in the nucleus. Recent studies suggest the role of Fhl2 in tissue repair and the anti-inflammatory response. Herein, we show that Fhl2-deficient mice develop a more severe psoriatic arthritis disease under induction of the inducible human tumor necrosis factor (hTNF) transgene than wild-type mice. The disease was accompanied by increased infiltration of activated macrophages and T regulatory cells in skin and digit joints as well as by increased expression of matrix metalloproteases and bone-specific proteases. The more severe pathogenesis of psoriatic arthritis in Fhl2 knockout mice coincided with enhanced levels of soluble hTNF cytokine, but surprisingly not with transcription of the hTNF transgene. Studying the shedding of cell membrane-bound hTNF by Adam17, a known Fhl2 interacting protein, revealed an enhanced release of TNF in the absence of Fhl2. In summary, our results show that Fhl2 anticipates the emerging inflammation and specifically the development of psoriatic arthritis by impeding the Adam17-mediated release of TNF.


Asunto(s)
Proteína ADAM17/metabolismo , Artritis Psoriásica/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Factores de Transcripción/metabolismo , Animales , Artritis Psoriásica/genética , Células Cultivadas , Proteínas de Homeodominio/metabolismo , Humanos , Inflamación/metabolismo , Proteínas con Homeodominio LIM/genética , Ratones Noqueados , Proteínas Musculares/genética , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Pathol ; 240(3): 366-377, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27555499

RESUMEN

We recently described an inducible human TNF transgenic mouse line (ihTNFtg) that develops psoriasis-like arthritis after doxycycline stimulation and analysed the pathogenesis of arthritis in detail. Here, we show that the skin phenotype of these mice is characterized by hyperproliferation and aberrant activation of keratinocytes, induction of pro-inflammatory cytokines, and infiltration with Th1 and Treg lymphocytes, particularly with macrophage infiltration into lesional skin, thus pointing to a psoriasis-like phenotype. To reveal the contribution of T cells and macrophages to the development of TNF-mediated psoriasis, ihTNFtg mice were crossbred into RAG1KO mice lacking mature T and B cells. Surprisingly, the psoriatic phenotype in the double mutants was not reduced; rather, it was enhanced. The skin showed significantly increased inflammation and in particular, increased infiltration by macrophages. Consequently, depletion of macrophages in RAG1KO or wild-type mice led to decreased disease severity. On the contrary, depletion of Treg cells in wild-type mice increased both psoriasis and the number of infiltrating macrophages, while adoptive transfer of Foxp3-positive cells into RAG1KO or wild-type mice decreased both the development of psoriasis and macrophage infiltration. Thus, we conclude that Treg lymphocytes inhibit the pro-inflammatory activity of macrophages, which are the major immune effector cells in hTNF-mediated psoriasis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Factores de Transcripción Forkhead/genética , Proteínas de Homeodominio/genética , Macrófagos/inmunología , Psoriasis/inmunología , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/genética , Traslado Adoptivo , Animales , Microambiente Celular , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Inflamación/patología , Queratinocitos/inmunología , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Psoriasis/patología , Piel/patología , Factor de Necrosis Tumoral alfa/metabolismo
6.
PLoS One ; 8(11): e81356, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260575

RESUMEN

Fibrogenesis is usually initiated when regenerative processes have failed and/or chronic inflammation occurs. It is characterised by the activation of tissue fibroblasts and dysregulated synthesis of extracellular matrix proteins. FHL2 (four-and-a-half LIM domain protein 2) is a scaffolding protein that interacts with numerous cellular proteins, regulating signalling cascades and gene transcription. It is involved in tissue remodelling and tumour progression. Recent data suggest that FHL2 might support fibrogenesis by maintaining the transcriptional expression of alpha smooth muscle actin and the excessive synthesis and assembly of matrix proteins in activated fibroblasts. Here, we present evidence that FHL2 does not promote bleomycin-induced lung fibrosis, but rather suppresses this process by attenuating lung inflammation. Loss of FHL2 results in increased expression of the pro-inflammatory matrix protein tenascin C and downregulation of the macrophage activating C-type lectin receptor DC-SIGN. Consequently, FHL2 knockout mice developed a severe and long-lasting lung pathology following bleomycin administration due to enhanced expression of tenascin C and impaired activation of inflammation-resolving macrophages.


Asunto(s)
Fibroblastos/metabolismo , Proteínas con Homeodominio LIM/genética , Macrófagos Peritoneales/patología , Proteínas Musculares/genética , Neumonía/genética , Fibrosis Pulmonar/genética , Tenascina/genética , Factores de Transcripción/genética , Animales , Bleomicina , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/patología , Regulación de la Expresión Génica , Proteínas con Homeodominio LIM/antagonistas & inhibidores , Proteínas con Homeodominio LIM/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación de Macrófagos , Macrófagos Peritoneales/metabolismo , Ratones , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Tenascina/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transcripción Genética
7.
Arthritis Rheum ; 65(9): 2290-300, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23740547

RESUMEN

OBJECTIVE: To generate doxycycline-inducible human tumor necrosis factor α (TNFα)-transgenic mice to overcome a major disadvantage of existing transgenic mice with constitutive expression of TNFα, which is the limitation in crossing them with various knockout or transgenic mice. METHODS: A transgenic mouse line that expresses the human TNFα cytokine exclusively after doxycycline administration was generated and analyzed for the onset of diseases. RESULTS: Doxycycline-inducible human TNFα-transgenic mice developed an inflammatory arthritis- and psoriasis-like phenotype, with fore and hind paws being prominently affected. The formation of "sausage digits" with characteristic involvement of the distal interphalangeal joints and nail malformation was observed. Synovial hyperplasia, enthesitis, cartilage and bone alterations, formation of pannus tissue, and inflammation of the skin epidermis and nail matrix appeared as early as 1 week after the treatment of mice with doxycycline and became aggravated over time. The abrogation of human TNFα expression by the removal of doxycycline 6 weeks after beginning stimulation resulted in fast resolution of the most advanced macroscopic and histologic disorders, and 3-6 weeks later, only minimal signs of disease were visible. CONCLUSION: Upon doxycycline administration, the doxycycline-inducible human TNFα-transgenic mouse displays the major features of inflammatory arthritis. It represents a unique animal model for studying the molecular mechanisms of arthritis, especially the early phases of disease genesis and tissue remodeling steps upon abrogation of TNFα expression. Furthermore, unlimited crossing of doxycycline-inducible human TNFα-transgenic mice with various knockout or transgenic mice opens new possibilities for unraveling the role of various signaling molecules acting in concert with TNFα.


Asunto(s)
Artritis Experimental/genética , Artritis Psoriásica/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Artritis Psoriásica/metabolismo , Artritis Psoriásica/patología , Cartílago/metabolismo , Cartílago/patología , Inflamación/patología , Articulaciones/metabolismo , Articulaciones/patología , Ratones , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Virol ; 85(5): 2333-41, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21159877

RESUMEN

Most retroviruses express all of their genes from a single primary transcript. In order to allow expression of more than one gene from this RNA, differential splicing is extensively used. Cellular quality control mechanisms retain and degrade unspliced or partially spliced RNAs in the nucleus. Two pathways have been described that explain how retroviruses circumvent this nuclear export inhibition. One involves a constitutive transport element in the viral RNA that interacts with the cellular mRNA transporter proteins NXF1 and NXT1 to facilitate nuclear export. The other pathway relies on the recognition of a viral RNA element by a virus-encoded protein that interacts with the karyopherin CRM1. In this report, we analyze the protein factors required for the nuclear export of unspliced foamy virus (FV) mRNA. We show that this export is CRM1 dependent. In contrast to other complex retroviruses, FVs do not encode an export-mediating protein. Cross-linking experiments indicated that the cellular protein HuR binds to the FV RNA. Inhibition studies showed that both ANP32A and ANP32B, which are known to bridge HuR and CRM1, are essential for FV RNA export. By using this export pathway, FVs solve a central problem of viral replication.


Asunto(s)
Núcleo Celular/metabolismo , ARN Viral/metabolismo , Infecciones por Retroviridae/virología , Spumavirus/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/genética , Células HEK293 , Humanos , Carioferinas/metabolismo , Transporte de ARN , ARN Viral/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Infecciones por Retroviridae/metabolismo , Spumavirus/genética , Proteína Exportina 1
9.
Med Microbiol Immunol ; 197(4): 381-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18351388

RESUMEN

We have isolated a Chikungunya (Chik) virus from a patient who returned to Germany after a three-month visit to Mauritius in spring 2006. Upon return she developed a transient fever up to 40 degrees C. This was followed by myalgia and joint pain. IgG antibodies in serum to Chik virus were undetectable. Virus (Chik-Wü1) was isolated on Vero cells. We molecularly cloned the whole genome of Chik-Wü1 from viral RNA by RT-PCR. The complete sequence was determined and functional domains of the genome were assigned. Chik-Wü1 clearly belongs to the group of viruses analysed from the recent Indian Ocean outbreak. In order to develop tools useful for further characterization of Chik-Wü1, we bacterially expressed and purified the capsid (C) and envelope (E) proteins and established an immunoblot assay. Twenty-two of 30 serum samples from Chik virus-infected patients that scored positive in indirect immunofluorescence previously were also reactive in immunoblot analysis with recombinant C and E2 antigens.


Asunto(s)
Infecciones por Alphavirus/virología , Virus Chikungunya/aislamiento & purificación , Genoma Viral , Immunoblotting/métodos , Adulto , Infecciones por Alphavirus/diagnóstico , Animales , Proteínas de la Cápside/genética , Virus Chikungunya/genética , Chlorocebus aethiops , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Mauricio , ARN Viral/genética , Análisis de Secuencia de ARN , Viaje , Células Vero , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA