Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(5): 1002-1021.e10, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33761330

RESUMEN

Arthritis typically involves recurrence and progressive worsening at specific predilection sites, but the checkpoints between remission and persistence remain unknown. Here, we defined the molecular and cellular mechanisms of this inflammation-mediated tissue priming. Re-exposure to inflammatory stimuli caused aggravated arthritis in rodent models. Tissue priming developed locally and independently of adaptive immunity. Repeatedly stimulated primed synovial fibroblasts (SFs) exhibited enhanced metabolic activity inducing functional changes with intensified migration, invasiveness and osteoclastogenesis. Meanwhile, human SF from patients with established arthritis displayed a similar primed phenotype. Transcriptomic and epigenomic analyses as well as genetic and pharmacological targeting demonstrated that inflammatory tissue priming relies on intracellular complement C3- and C3a receptor-activation and downstream mammalian target of rapamycin- and hypoxia-inducible factor 1α-mediated metabolic SF invigoration that prevents activation-induced senescence, enhances NLRP3 inflammasome activity, and in consequence sensitizes tissue for inflammation. Our study suggests possibilities for therapeutic intervention abrogating tissue priming without immunosuppression.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Fibroblastos/inmunología , Inflamación/inmunología , Membrana Sinovial/inmunología , Inmunidad Adaptativa/inmunología , Animales , Artritis Reumatoide/inmunología , Línea Celular , Perros , Humanos , Mediadores de Inflamación/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratas Wistar , Transducción de Señal/inmunología
2.
Cell Commun Signal ; 22(1): 281, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773618

RESUMEN

BACKGROUND: Restoring impaired peripheral immune tolerance is the primary challenge in treating autoimmune diseases. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs), a fraction of low molecular weight proteins, in inhibiting the progression of psoriatic arthritis, even in the presence of high levels of the proinflammatory cytokine TNFα in the bloodstream. When specifically targeting dendritic cells (DCs), SSPs transform them into tolerogenic cells, which efficiently induce the development of regulatory Foxp3+ Treg cells. In this study, we provide further insights into the mechanism of action of SSPs. RESULTS: We found that SSPs stimulate the activation of the mTOR signaling pathway in dendritic cells, albeit in a different manner than the classical immunogenic stimulus LPS. While LPS-induced activation is rapid, strong, and sustained, the activity induced by SSPs is delayed, less intense, yet still significant. These distinct patterns of activation, as measured by phosphorylation of key components of the pathway are also observed in response to other immunogenic and tolerogenic stimuli such as GM-CSF + IL-4 or IL-10 and TGFß. The disparity in mTOR activation between immunogenic and tolerogenic stimuli is quantitative rather than qualitative. In both cases, mTOR activation primarily occurs through the PI3K/Akt signaling axis and involves ERK and GSK3ß kinases, with minimal involvement of AMPK or NF-kB pathways. Furthermore, in the case of SSPs, mTOR activation seems to involve adenosine receptors. Additionally, we observed that DCs treated with SSPs exhibit an energy metabolism with high plasticity, which is typical of tolerogenic cells rather than immunogenic cells. CONCLUSION: Hence, the decision whether dendritic cells enter an inflammatory or tolerogenic state seems to rely on varying activation thresholds and kinetics of the mTOR signaling pathway.


Asunto(s)
Células Dendríticas , Tolerancia Inmunológica , Transducción de Señal , Serina-Treonina Quinasas TOR , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Ratones , Inflamación/metabolismo , Cinética , Lipopolisacáridos/farmacología
3.
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
4.
J Virol ; 95(6)2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33408177

RESUMEN

Influenza A virus (IAV) nonstructural protein 1 (NS1) is a protein with multiple functions that are regulated by phosphorylation. Phosphoproteomic screening of H1N1 virus-infected cells revealed that NS1 was phosphorylated at serine 205 in intermediate stages of the viral life cycle. Interestingly, S205 is one of six amino acid changes in NS1 of post-pandemic H1N1 viruses currently circulating in humans compared to the original swine-origin 2009 pandemic (H1N1pdm09) virus, suggesting a role in host adaptation. To identify NS1 functions regulated by S205 phosphorylation, we generated recombinant PR8 H1N1 NS1 mutants with S205G (nonphosphorylatable) or S205N (H1N1pdm09 signature), as well as H1N1pdm09 viruses harboring the reverse mutation NS1 N205S or N205D (phosphomimetic). Replication of PR8 NS1 mutants was attenuated relative to wild-type (WT) virus replication in a porcine cell line. However, PR8 NS1 S205N showed remarkably higher attenuation than PR8 NS1 S205G in a human cell line, highlighting a potential host-independent advantage of phosphorylatable S205, while an asparagine at this position led to a potential host-specific attenuation. Interestingly, PR8 NS1 S205G did not show polymerase activity-enhancing functions, in contrast to the WT, which can be attributed to diminished interaction with cellular restriction factor DDX21. Analysis of the respective kinase mediating S205 phosphorylation indicated an involvement of casein kinase 2 (CK2). CK2 inhibition significantly reduced the replication of WT viruses and decreased NS1-DDX21 interaction, as observed for NS1 S205G. In summary, NS1 S205 is required for efficient NS1-DDX21 binding, resulting in enhanced viral polymerase activity, which is likely to be regulated by transient phosphorylation.IMPORTANCE Influenza A viruses (IAVs) still pose a major threat to human health worldwide. As a zoonotic virus, IAV can spontaneously overcome species barriers and even reside in new hosts after efficient adaptation. Investigation of the functions of specific adaptational mutations can lead to a deeper understanding of viral replication in specific hosts and can probably help to find new targets for antiviral intervention. In the present study, we analyzed the role of NS1 S205, a phosphorylation site that was reacquired during the circulation of pandemic H1N1pdm09 "swine flu" in the human host. We found that phosphorylation of human H1N1 virus NS1 S205 is mediated by the cellular kinase CK2 and is needed for efficient interaction with human host restriction factor DDX21, mediating NS1-induced enhancement of viral polymerase activity. Therefore, targeting CK2 activity might be an efficient strategy for limiting the replication of IAVs circulating in the human population.


Asunto(s)
Virus de la Influenza A/fisiología , ARN Polimerasa Dependiente del ARN/metabolismo , Serina/metabolismo , Proteínas no Estructurales Virales/metabolismo , Adaptación Fisiológica/genética , Animales , Quinasa de la Caseína II/metabolismo , Línea Celular , ARN Helicasas DEAD-box/metabolismo , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Mutación , Fosforilación , Unión Proteica , Porcinos , Proteínas no Estructurales Virales/genética , Replicación Viral
5.
J Virol ; 95(20): e0067221, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34319782

RESUMEN

Influenza A virus (IAV) is the causative agent of flu disease that results in annual epidemics and occasional pandemics. IAV alters several signaling pathways of the cellular host response in order to promote its replication. Therefore, some of these pathways can serve as targets for novel antiviral agents. Here, we show that c-Jun NH2-terminal kinase (JNK)-interacting protein 4 (JIP4) is dynamically phosphorylated in IAV infection. The lack of JIP4 resulted in higher virus titers, with significant differences in viral protein and mRNA accumulation as early as within the first replication cycle. In accordance, decreased IAV titers and protein accumulation were observed during the overexpression of JIP4. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 mitogen-activated protein kinase (MAPK) pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a direct reduction of viral polymerase activity. Furthermore, the interference of JIP4 with IAV replication seems to be linked to the phosphorylation of the serine at position 730 that is sufficient to impede the viral polymerase. Collectively, we provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730. IMPORTANCE Influenza A virus (IAV) infection is a world health concern, and current treatment options encounter high rates of resistance. Our group investigates host pathways modified in IAV infection as promising new targets. The host protein JIP4 is dynamically phosphorylated in IAV infection. JIP4 absence resulted in higher virus titers and viral protein and mRNA accumulation within the first replication cycle. Accordingly, decreased IAV titers and protein accumulation were observed during JIP4 overexpression. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 MAPK pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a reduction in viral polymerase activity. The interference of JIP4 with IAV replication is linked to the phosphorylation of serine 730. We provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus de la Influenza A/metabolismo , Células A549 , Animales , Chlorocebus aethiops , Perros , Interacciones Huésped-Patógeno/genética , Humanos , Evasión Inmune/genética , Inmunidad Innata/genética , Virus de la Influenza A/fisiología , Gripe Humana/virología , Interferones/genética , Células de Riñón Canino Madin Darby , Fosforilación , Transducción de Señal/genética , Células Vero , Replicación Viral/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Cell Sci ; 132(6)2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30745335

RESUMEN

The four and a half LIM domains protein 2 (Fhl2) is an adaptor protein capable of mediating protein-protein interactions. Here, we report for the first time phenotypic changes in the brain of Fhl2-deficient mice. We showed that Fhl2 is expressed in neural stem cells, precursors and mature cells of neuronal lineage. Moreover, Fhl2 deficiency leads to delayed neuroblast migration in vivo, premature astroglial differentiation of neural stem cells (NSCs) in vitro, and a gliosis-like accumulation of glial fibrillary acidic protein (GFAP)-positive astrocytes in vivo that substantially increases with age. Collectively, Fhl2-deficiency in the brain interrupts the maintenance and the balanced differentiation of adult NSCs, resulting in preferentially glial differentiation and early exhaustion of the NSC pool required for adult neurogenesis.


Asunto(s)
Movimiento Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Factores de Transcripción/metabolismo , Envejecimiento , Animales , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular , Células Cultivadas , Gliosis/metabolismo , Células HEK293 , Humanos , Ratones , Células-Madre Neurales/citología , Neuronas/metabolismo
7.
J Pathol ; 251(4): 388-399, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32449525

RESUMEN

Recently, we established a doxycycline-inducible human tumor necrosis factor alpha (TNFα)-transgenic mouse line, ihTNFtg. Non-induced young and elderly mice showed low but constitutive expression of hTNFα due to promoter leakiness. The persistently present hTNFα stimulated endogenous pro-inflammatory mouse mS100A8/A9 alarmins. Secreted mS100A8/A9 in turn induced the expression and release of mouse mTNFα. The continuous upregulation of pro-inflammatory mTNFα and mS100A8/A9 proteins, due to their mutual expression dependency, gradually led to increased levels in colon tissue and blood. This finally exceeded the threshold levels tolerated by the healthy organism, leading to the onset of intestinal inflammation. Here, recombinant hTNFα functioned as an initial trigger for the development of chronic inflammation. Crossing ihTNFtg mice with S100A9KO mice lacking active S100A8/A9 alarmins or with Rag1KO mice lacking T and B lymphocytes completely abrogated the development of colonic inflammation, despite the still leaky hTNFα promoter. Furthermore, both the intensity of the immune response and the strength of immunosuppressive Treg induction was found to depend on the major histocompatibility complex (MHC) genetic composition. In summary, the onset of intestinal inflammation in elderly mice depends on at least four factors that have to be present simultaneously: TNFα upregulation, S100A8/A9 protein expression, functional T lymphocytes and genetic composition, with the MHC haplotype being of central importance. Only joint action of these factors leads to chronic intestinal inflammation, while absence of any of these determinants abrogates the development of the autoimmune disorder. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Colitis/genética , Inflamación/genética , Complejo Mayor de Histocompatibilidad/genética , Factor de Necrosis Tumoral alfa/metabolismo , Alarminas/genética , Alarminas/metabolismo , Animales , Células de la Médula Ósea , Calgranulina A/genética , Calgranulina B/genética , Colitis/metabolismo , Colitis/patología , Colon/metabolismo , Colon/patología , Haplotipos , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
8.
FASEB J ; 33(7): 7799-7809, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30939249

RESUMEN

The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.


Asunto(s)
Inflamación/fisiopatología , Proteínas con Homeodominio LIM/fisiología , Proteínas Musculares/fisiología , Factores de Transcripción/fisiología , Cicatrización de Heridas/fisiología , Animales , Quimiotaxis de Leucocito/fisiología , Citocinas/fisiología , Transición Epitelial-Mesenquimal/fisiología , Inflamación/inmunología , Proteínas con Homeodominio LIM/biosíntesis , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Musculares/biosíntesis , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Miofibroblastos/fisiología , Regeneración/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética/fisiología , Regulación hacia Arriba
9.
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
10.
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
11.
Cell Microbiol ; 19(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28076660

RESUMEN

Expression of the influenza A virus (IAV) nonstructural protein (NS1) results in the activation of c-Jun N-terminal kinase (JNK). Both NS1 and JNK are involved in apoptosis induction. To investigate their interrelationship, we stably expressed a tamoxifen inducible NS1 oestrogen receptor fusion-protein (NS1ERT) in mammalian cells. Upon tamoxifen stimulation, NS1ERT-expressing cells partially rescued the attenuated replication of NS1-deficient IAVs and also inhibited interferon up-regulation, confirming the functional competence of NS1ERT. Tamoxifen-induced NS1ERT created a cytopathic phenotype and led to the activation of JNK and apoptosis. Induction of NS1F103SERT mutant failed to activate JNK, but induced apoptosis, whereas the induction of NS1M106IERT led to JNK phosphorylation, but not apoptosis, indicating that JNK activation and apoptosis induction are not functionally linked. Further mutational analysis highlighted that apoptosis induction is a function of the C-terminal effector domain of NS1. Finally, IAVs encoding mutant NS1 revealed a modulating effect of NS1 on apoptosis induction in a genuine infection. With respect to apoptogenicity, an NS1 mutant virus that results in a super activation of JNK behaves similarly to the JNK nonactivating virus expressing NS1F103S, thus confirming that NS1-mediated JNK activation and apoptosis induction are also functionally independent from each other in vivo.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas no Estructurales Virales/genética , Células A549 , Animales , Línea Celular , Perros , Activación Enzimática , Células HEK293 , Humanos , Virus de la Influenza A/metabolismo , Células de Riñón Canino Madin Darby , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tamoxifeno/metabolismo , Proteínas no Estructurales Virales/metabolismo
12.
Cell Microbiol ; 18(6): 784-91, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26687707

RESUMEN

Phosphorylation and dephosphorylation acts as a fundamental molecular switch that alters protein function and thereby regulates many cellular processes. The non-structural protein 1 (NS1) of influenza A virus is an important factor regulating virulence by counteracting cellular immune responses against viral infection. NS1 was shown to be phosphorylated at several sites; however, so far, no function has been conclusively assigned to these post-translational events yet. Here, we show that the newly identified phospho-site threonine 49 of NS1 is differentially phosphorylated in the viral replication cycle. Phosphorylation impairs binding of NS1 to double-stranded RNA and TRIM25 as well as complex formation with RIG-I, thereby switching off its interferon antagonistic activity. Because phosphorylation was shown to occur at later stages of infection, we hypothesize that at this stage other functions of the multifunctional NS1 beyond its interferon-antagonistic activity are needed.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Interferón beta/metabolismo , Treonina/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box/metabolismo , Perros , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Interferón beta/genética , Células de Riñón Canino Madin Darby/virología , Mutación , Fosforilación , Regiones Promotoras Genéticas , Receptores Inmunológicos , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral
13.
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
14.
Ann Rheum Dis ; 74(12): 2216-23, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25125695

RESUMEN

OBJECTIVE: We analysed the role of the adaptor molecule four-and-a-half Lin11, Isl-1 & Mec-3 (LIM) domain protein 2 (FHL2) in the activation of fibroblast-like synoviocytes in human rheumatoid arthritis (RA) and tumour necrosis factor α (TNFα)-dependent animal models of the disease. METHODS: Synovial tissues of patients with RA and osteoarthritis (OA) as well as hind paw sections from arthritic human TNFα transgenic (hTNFtg) mice and synovial fibroblasts from these were analysed. The effects of cytokines on the expression of FHL2 and disease-relevant matrixmetalloproteases (MMPs) were determined. Analyses of human tissue specimens from patients treated with anti-TNFα as well as anti-TNFα treatment of hTNFtg mice were performed to substantiate the TNFα effects on FHL2 levels. FHL2(-/-) mice and hTNFtg mice (with constitutive or inducible transgene expression) were crossbred to generate TNFα overexpressing FHL2-deficient animals. Signalling pathways were analysed in cells from these mice and in human cells after knock down of FHL2 by western blot. RESULTS: FHL2 levels were higher in RA than in OA and in hTNFtg than in wild-type mice. Surprisingly, while transforming growth factor (TGF)ß-induced FHL2 expression, TNFα suppressed FHL2. In vivo, anti-TNFα treatment led to higher FHL2 levels both in RA patients and hTNFtg mice. The loss of FHL2 increased joint destruction in hTNFtg mice, which was accompanied by elevated MMP-13. In vitro, TNFα-mediated MMP-13 was significantly higher in FHL2(-/-) cells and after knock down of FHL2, which was caused by prolonged p38 MAPK activation. CONCLUSIONS: These data suggest that FHL2 serves as a protective factor and that, rather than promoting the pathology, the upregulation of FHL2 in RA occurs in frame of a regenerative attempt.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/genética , Osteoartritis/genética , Membrana Sinovial/metabolismo , Factores de Transcripción/genética , Animales , Células Cultivadas , Enfermedad Crónica , Humanos , Immunoblotting , Proteínas con Homeodominio LIM/biosíntesis , Ratones , Ratones Transgénicos , Proteínas Musculares/biosíntesis , Osteoartritis/metabolismo , Osteoartritis/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Membrana Sinovial/patología , Factores de Transcripción/biosíntesis
15.
FASEB J ; 28(10): 4235-46, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24935968

RESUMEN

MK2 and MK3 are downstream targets of p38 and ERK1/2. They control the mRNA stability of several inflammatory cytokines, including TNF-α and IL-10. Whereas MK2 is expressed ubiquitously, the expression of MK3 is restricted to muscle, liver, and heart tissues and T and NK cells. Using Mk-deficient and wild-type (WT) mice, we demonstrated an inhibitory effect of MK3, but not of MK2, on interferon (IFN)-γ expression in T and NK lymphocytes. The results provided evidence that the inhibitory effect of MK3 is based on negative feedback phosphorylation of p38 and ERK1/2, which causes decreased binding of Stat4 to the IFN-γ promoter and reduced expression of IFN-γ mRNA and protein. Consequently, all Mk3(-/-) mice challenged with the Th1-inducing influenza A virus (IAV) survived the WT LD50 virus dose. The reduced disease severity in the Mk3(-/-) mice was accompanied by a >10-fold reduction in viral lung titer and an increase in the number of activated NK cells and enhanced Th1 activation of CD4 T cells. Thus, our data describe the protein kinase MK3 as a novel regulator of the innate and adaptive immune responses.-Köther, K., Nordhoff, C., Masemann, D., Varga, G., Bream, J. H., Gaestel, M., Wixler, V., Ludwig, S. MAPKAP kinase 3 suppresses Ifng gene expression and attenuates NK cell cytotoxicity and Th1 CD4 T-cell development upon influenza A virus infection.


Asunto(s)
Citotoxicidad Inmunológica , Virus de la Influenza A , Interferón gamma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Células TH1/inmunología , Animales , Regulación de la Expresión Génica , Interferón gamma/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Activación de Linfocitos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo
16.
Cell Commun Signal ; 12: 29, 2014 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-24767605

RESUMEN

BACKGROUND: The replication cycle of most pathogens, including influenza viruses, is perfectly adapted to the metabolism and signal transduction pathways of host cells. After infection, influenza viruses activate several cellular signaling cascades that support their propagation but suppress those that interfere with viral replication. Accumulation of viral RNA plays thereby a central role. Its sensing by the pattern recognition receptors of the host cells leads to the activation of several signal transduction waves that result in induction of genes, responsible for the cellular innate immune response. Type I interferon (IFN) genes and interferon-stimulated genes (ISG) coding for antiviral-acting proteins, such as MxA, OAS-1 or PKR, are primary targets of these signaling cascades. ß- and γ-catenin are closely related armadillo repeat-containing proteins with dual roles. At the cell membrane they serve as adapter molecules linking cell-cell contacts to microfilaments. In the cytosol and nucleus, the proteins form a transcriptional complex with the lymphoid enhancer factor/T-cell factor (LEF/TCF), regulating the transcription of many genes, thereby controlling different cellular functions such as cell cycle progression and differentiation. RESULTS: In this study, we demonstrate that ß- and γ-catenin are important regulators of the innate cellular immune response to influenza A virus (IAV) infections. They inhibit viral replication in lung epithelial cells by enhancing the virus-dependent induction of the IFNB1 gene and interferon-stimulated genes. Simultaneously, the prolonged infection counteracts the antiviral effect of ß- and γ-catenin. Influenza viruses suppress ß-catenin-dependent transcription by misusing the RIG-I/NF-κB signaling cascade that is induced in the course of infection by viral RNA. CONCLUSION: We identified ß- and γ-catenin as novel antiviral-acting proteins. While these factors support the induction of common target genes of the cellular innate immune response, their functional activity is suppressed by pathogen evasion.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Interferón beta/metabolismo , FN-kappa B/metabolismo , Receptores de Ácido Retinoico/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Perros , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células HEK293 , Humanos , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/fisiología , Interferón beta/genética , Pulmón/citología , Células de Riñón Canino Madin Darby , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Células Vero , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral , gamma Catenina/metabolismo
17.
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
18.
Biomolecules ; 14(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38672485

RESUMEN

Restoring peripheral immune tolerance is crucial for addressing autoimmune diseases. An ancient mechanism in maintaining the balance between inflammation and tolerance is the ratio of extracellular ATP (exATP) and adenosine. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs) in inhibiting psoriatic arthritis progression, even in the presence of the pro-inflammatory cytokine TNFα, by transforming dendritic cells (DCs) into tolerogenic cells and fostering regulatory Foxp3+ Treg cells. Here, we identified thymosins as the primary constituents of SSPs, but recombinant thymosin peptides were less efficient in inhibiting arthritis than SSPs. Since Tß4 is an ecto-ATPase-binding protein, we hypothesized that SSPs regulate exATP profiles. Real-time investigation of exATP levels in DCs revealed that tolerogenic stimulation led to robust de novo exATP synthesis followed by significant degradation, while immunogenic stimulation resulted in a less pronounced increase in exATP and less effective degradation. These contrasting exATP profiles were crucial in determining whether DCs entered an inflammatory or tolerogenic state, highlighting the significance of SSPs as natural regulators of peripheral immunological tolerance, with potential therapeutic benefits for autoimmune diseases. Finally, we demonstrated that the tolerogenic phenotype of SSPs is mainly influenced by adenosine receptors, and in vivo administration of SSPs inhibits psoriatic skin inflammation.


Asunto(s)
Adenosina Trifosfato , Diferenciación Celular , Células Dendríticas , Bazo , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Bazo/citología , Bazo/metabolismo , Bazo/efectos de los fármacos , Bazo/inmunología , Ratones , Timosina/farmacología , Timosina/metabolismo , Péptidos/farmacología , Artritis Psoriásica/tratamiento farmacológico , Artritis Psoriásica/metabolismo , Artritis Psoriásica/inmunología , Humanos , Ratones Endogámicos C57BL , Tolerancia Inmunológica/efectos de los fármacos
19.
FASEB J ; 26(3): 1290-300, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22106366

RESUMEN

Guanylate-binding proteins (GBPs) belong to the family of large GTPases that are induced in response to interferons. GBPs contain an N-terminal globular GTPase domain and a C-terminal α-helical regulatory domain that are connected by a short middle domain. Antiviral activity against vesicular stomatitis virus and encephalomyocarditis virus has been shown for hGBP-1; however, no anti-influenza virus properties for GBPs have been described to date. Here we show that hGBP-1 and hGBP-3 possess anti-influenza viral activity. Furthermore, we have identified a novel splice variant of hGBP-3, named hGBP-3ΔC, with a largely modified C-terminal α-helical domain. While all three GBP isoforms were up-regulated on influenza virus infection, hGBP-3ΔC showed the most prominent antiviral activity in epithelial cells. Mutational analysis of hGBPs revealed that the globular domain is the principal antiviral effector domain, and GTP-binding, but not hydrolysis, is necessary for antiviral action. Furthermore, we showed that hGBP-3ΔC strongly represses the activity of the viral polymerase complex, which results in decreased synthesis of viral vRNA, cRNA, mRNA, and viral proteins, as well.


Asunto(s)
Proteínas de Unión al GTP/genética , Transcripción Genética , Replicación Viral , Virus/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Línea Celular , Línea Celular Tumoral , Citocinas/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Proteínas de Unión al GTP/inmunología , Proteínas de Unión al GTP/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Datos de Secuencia Molecular , Mutación , Orthomyxoviridae/genética , Orthomyxoviridae/inmunología , Orthomyxoviridae/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Virus de la Estomatitis Vesicular Indiana/fisiología , Virus/inmunología
20.
Cell Microbiol ; 14(7): 1135-47, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22417706

RESUMEN

The innate immune response of influenza A virus-infected cells is predominantly mediated by type I interferon-induced proteins. Expression of the interferon ß (IFNß) itself is initiated by accumulating viral RNA and is transmitted by different signalling cascades that feed into activation of the three transcriptional elements located in the IFNß promoter, AP-1, IRF-3 and NF-κB. FHL2 (four-and-a-half LIM domain protein 2) is an adaptor molecule that shuttles between membrane and nucleus regulating signalling cascades and gene transcription. Here we describe FHL2 as a novel regulator of influenza A virus propagation. Using mouse FHL2 wild-type, knockout and rescued cells and human epithelial cells with different expression levels of FHL2 we showed that FHL2 decreases influenza A virus propagation by regulating the intrinsic cellular antiviral immune response. On virus infection FHL2 translocates into the nucleus, potentiating the IRF-3-dependent transcription of the IFNß gene.


Asunto(s)
Inmunidad Innata , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/biosíntesis , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA