Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Biomed Pharmacother ; 171: 116153, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38232664

RESUMEN

Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory disease characterized by persistent inflammation and joint destruction. A lipid mediator (LM, namely, 17S-monohydroxy docosahexaenoic acid, resolvin D5, and protectin DX in a ratio of 3:47:50) produced by soybean lipoxygenase from DHA, exhibits anti-inflammatory activity. In this study, we determined the effect of LM on collagen antibody-induced arthritis (CAIA) in mice and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation in RAW264.7 cells. LM effectively downregulated the expression of tartrate-resistant acid phosphatase (TRAP) and cathepsin K, inhibited osteoclast formation, and suppressed the NF-κB signaling pathway in vitro. In vivo, LM at 10 µg/kg/day significantly decreased paw swelling and inhibited progression of arthritis in CAIA mice. Moreover, proinflammatory cytokine (tumor necrosis factor-α, interleukin (IL)-6, IL-1ß, IL-17, and interferon-γ) levels in serum were decreased, whereas IL-10 levels were increased following LM treatment. Furthermore, LM alleviated joint inflammation, cartilage erosion, and bone destruction in the ankles, which may be related to matrix metalloproteinase and Janus kinase (JAK)-signal transducer and activators of transcription (STAT) signaling pathway. Our findings suggest that LM attenuates arthritis severity, restores serum imbalances, and modifies joint damage. Thus, LM represents a promising therapy for relieving RA symptoms.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Ratones , Animales , Osteoclastos , Ligando RANK/metabolismo , Glycine max , Ácidos Docosahexaenoicos/farmacología , Artritis Reumatoide/metabolismo , Artritis Experimental/patología , Inflamación/metabolismo , Lipooxigenasas/metabolismo , Lipooxigenasas/farmacología
3.
Mol Ther Oncolytics ; 30: 301-315, 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37727704

RESUMEN

Oncolytic viruses are of significant clinical interest due to their ability to directly infect and kill tumors and enhance the anti-tumor immune response. Previously, we developed KLS-3010, a novel oncolytic virus derived from the International Health Department-White (IHD-W) strain vaccinia virus, which has robust tumoricidal effects. In the present study, we generated a recombinant oncolytic virus, KLS-3020, by inserting three transgenes (hyaluronidase [PH-20], interleukin-12 [IL-12], and soluble programmed cell death 1 fused to the Fc domain [sPD1-Fc]) into KLS-3010 and investigated its anti-tumor efficacy and ability to induce anti-tumor immune responses in CT26.WT and B16F10 mouse tumor models. A single injection of KLS-3020 significantly decreased tumor growth. The roles of the transgenes were investigated using viruses expressing each single transgene alone and KLS-3020. PH-20 promoted virus spread and tumor immune cell infiltration, IL-12 activated and reprogrammed T cells to inflammatory phenotypes, and sPD1-Fc increased intra-tumoral populations of activated T cells. The tumor-specific systemic immune response and the abscopal tumor control elicited by KLS-3020 were demonstrated in the CT26.WT tumor model. The insertion of transgenes into KLS-3020 increased its anti-tumor efficacy, supporting further clinical investigation of KLS-3020 as a novel oncolytic immunotherapy.

4.
Int Immunopharmacol ; 124(Pt A): 110900, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37708704

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin condition that primarily results from immune dysregulation. We determined the potential therapeutic benefits of lipid mediators (LM, 17S-monohydroxy DHA, resolvin D5, and protectin DX in a ratio of 3:47:50) produced by soybean lipoxygenase from DHA. The underlying molecular mechanisms involved in TNF-α/IFN-γ-stimulated HaCaT cells as well as its effect in an AD mouse model induced by DNCB in BALB/c mice were examined. The results indicated that LM effectively attenuates the production of inflammatory cytokines (IL-6 and IL-1ß) and chemokines (IL-8 and MCP-1) by inhibiting the NF-κB signaling pathway in TNF-α/IFN-γ-stimulated HaCaT cells. The oral administration of LM at 5 or 10 µg/kg/day significantly reduced skin lesions, epidermal thickness, and mast cell infiltration in AD mice. Furthermore, LM reduced the production of IgE and inflammatory cytokines (TNF-α, IL-6, and IL-1ß) in the serum, modulated gut microbiota diversity, and restored the microbial composition. Overall, our findings suggest that LM represents a potential therapeutic agent for improving AD symptoms through its ability to suppress inflammatory cytokines and alter the composition of gut microbiota.

5.
Osteoarthritis Cartilage ; 31(12): 1567-1580, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37544583

RESUMEN

OBJECTIVE: TissueGene-C (TG-C), a combination of human allogeneic chondrocytes and irradiated GP2-293 cells engineered to overexpress transforming growth factor-ß1 (TGF-ß1), has been developed as a novel cell-based gene therapy and a candidate for disease modifying osteoarthritis drug (DMOAD). We aim to investigate analgesic mechanism of TG-C in a pre-clinical animal model with monoiodoacetate (MIA)-induced pain. DESIGN: We used a rat MIA model of osteoarthritis (OA) pain. We examined that TG-C can regulate pain by inhibiting the upregulation of various pain mediators in both knee joint tissue and dorsal root ganglia (DRG) (n = 112) and alleviating pain behavior (n = 41) and neuronal hyperexcitability in DRG (n = 60), afferent nerve fiber (n = 24), and spinal cord (n = 35). RESULTS: TG-C significantly alleviated pain-related behavior by restoring altered dynamic weight bearing and reduced mechanical threshold of the affected hindlimb. TG-C significantly suppressed the expression of nerve growth factor (NGF) and calcitonin gene-related peptide (CGRP) in inflamed joint tissue. TG-C significantly suppressed the upregulation of tropomyosin receptor kinase A (TrkA) and nerve injury/regeneration protein (GAP43) and activation of Iba1-positive microglial cells in DRG. TG-C significantly recovered neuronal hyperexcitability by restoring RMP and firing threshold and frequency of DRG neurons, attenuating firing rates of mechanosensitive C- or Aδ-nerve fiber innervating knee joint, and lowering increased miniature and evoked excitatory postsynaptic currents (mEPSCs and eEPSCs) in the spinal cord. CONCLUSION: Our results demonstrated that TG-C exerted potent analgesic effects in a rat MIA model of OA pain by inhibiting the upregulation of pain mediators and modulating neuronal sensitization.


Asunto(s)
Osteoartritis , Dolor , Ratas , Humanos , Animales , Ratas Sprague-Dawley , Dolor/metabolismo , Osteoartritis/terapia , Osteoartritis/tratamiento farmacológico , Analgésicos/uso terapéutico , Neuronas/metabolismo , Ganglios Espinales/metabolismo , Modelos Animales de Enfermedad
6.
J Pain ; 24(12): 2211-2227, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37442406

RESUMEN

Although peripheral neuropathic pain is caused by peripheral nerve injury, it is not simply a peripheral nervous system disease. It causes abnormalities in both the central and peripheral nervous systems. Pathological phenomena, such as hyperactivation of sensory neurons and inflammation, are observed in both the dorsal root ganglion and spinal cord. Pain signals originating from the periphery are transmitted to the brain via the SC, and the signals are modulated by pathologically changing SC conditions. Therefore, the modulation of SC pathology is important for peripheral NP treatment. We investigated the effects of KLS-2031 (recombinant adeno-associated viruses expressing glutamate decarboxylase 65, glial cell-derived neurotrophic factor, and interleukin-10) delivered to the dorsal root ganglion on aberrant neuronal excitability and neuroinflammation in the SC of rats with peripheral NP. Results showed that KLS-2031 administration restored excessive excitatory transmission and inhibitory signals in substantia gelatinosa neurons. Moreover, KLS-2031 restored the in vivo hypersensitivity of wide dynamic range neurons and mitigated neuroinflammation in the SC by regulating microglia and astrocytes. Collectively, these findings demonstrated that KLS-2031 efficiently suppressed pathological pain signals and inflammation in the SC of peripheral NP model, and is a potential novel therapeutic approach for NP in clinical settings. PERSPECTIVE: Our study demonstrated that KLS-2031, a combination gene therapy delivered by transforaminal epidural injection, not only mitigates neuroinflammation but also improves SC neurophysiological function, including excitatory-inhibitory balance. These findings support the potential of KLS-2031 as a novel modality that targets multiple aspects of the complex pathophysiology of neuropathic pain.


Asunto(s)
Neuralgia , Enfermedades Neuroinflamatorias , Ratas , Animales , Neuralgia/terapia , Médula Espinal , Terapia Genética , Inflamación , Células Receptoras Sensoriales , Hiperalgesia , Ganglios Espinales
7.
Am J Physiol Cell Physiol ; 325(1): C257-C271, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37306390

RESUMEN

Osteoarthritis (OA) is a currently incurable, chronic, progressive, and debilitating musculoskeletal (MSK) condition. One of its hallmark symptoms is chronic nociceptive and neuropathic pain, which significantly reduces the quality of life of patients with OA. Although research into the pathomechanisms of OA pain is ongoing and several pain pathways are well understood, the true source of OA pain remains unclear. Ion channels and transporters are key mediators of nociceptive pain. In this narrative review article, we summarize the state-of-the-art in relation to the distribution and function of ion channels in all major synovial joint tissues in the context of pain generation. We provide an update on the ion channels likely involved in mediating peripheral and central nociceptive pathways in the nervous system in OA pain, including voltage-gated sodium and potassium channels, members of the transient receptor potential (TRP) channel family, and purinergic receptor complexes. We focus on ion channels and transporters that have the potential to be candidate drug targets for pain management in patients with OA. We propose that ion channels expressed by the cells of constituent tissues of OA-afflicted synovial joints including cartilage, bone, synovium, ligament, and muscle, should be more thoroughly investigated and targeted in the context of OA pain. Based on key findings from recent basic research articles as well as clinical trials, we propose novel directions for the development of future analgesic therapies to improve the quality of life of patients with OA.


Asunto(s)
Osteoartritis , Canales de Potencial de Receptor Transitorio , Humanos , Calidad de Vida , Dolor/tratamiento farmacológico , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Inflamación
8.
Nutrients ; 15(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36678298

RESUMEN

Although fish oil (FO) and lipid mediators (LM) derived from polyunsaturated fatty acids can prevent obesity, their combined effects and cellular metabolism remain unclear. Therefore, this study aimed to examine the potential protective and metabolic effects of FO in combination with LM (a mixture of 17S-monohydroxy docosahexaenoic acid, resolvin D5, and protectin DX [3:47:50], derived from docosahexaenoic acid (DHA)) on palmitic acid (PA)-induced HepG2 cells and high-fat- diet (HFD)-induced C57BL/6J mice after 9-week treatment. Lipid metabolism disorders and inflammation induced by HFD and PA were substantially reduced after FO and LM treatment. Further, FO and LM treatments reduced lipid accumulation by increasing fatty acid oxidation via peroxisome proliferator-activated receptor α and carnitine-palmitoyl transferase 1 as well as by decreasing fatty acid synthesis via sterol regulatory element-binding protein-1c and fatty acid synthase. Finally, FO and LM treatment reduced inflammation by blocking the NF-κB signaling pathway. Importantly, the combination of FO and LM exhibited more robust efficacy against nonalcoholic fatty liver disease, suggesting that FO supplemented with LM is a beneficial dietary strategy for treating this disease.


Asunto(s)
Aceites de Pescado , Metabolismo de los Lípidos , Animales , Humanos , Ratones , Dieta Alta en Grasa , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/metabolismo , Aceites de Pescado/farmacología , Aceites de Pescado/metabolismo , Células Hep G2 , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL
9.
Ther Adv Musculoskelet Dis ; 14: 1759720X221085952, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36504595

RESUMEN

In this narrative review article, we critically assess the current state of the osteoarthritis (OA) drug development pipeline. We discuss the current state-of-the-art in relation to the development and evaluation of candidate disease-modifying OA drugs (DMOADs) and the limitations associated with the tools and methodologies that are used to assess outcomes in OA clinical trials. We focus on the definition of DMOADs, highlight the need for an updated definition in the form of a consensus statement from all the major stakeholders, including academia, industry, regulatory agencies, and patient organizations, and provide a summary of the results of recent clinical trials of novel DMOAD candidates. We propose that DMOADs should be more appropriately targeted and investigated according to the emerging clinical phenotypes and molecular endotypes of OA. Based on the findings from recent clinical trials, we propose key topics and directions for the development of future DMOADs.

10.
Hum Gene Ther ; 32(9-10): 517-527, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32854548

RESUMEN

Oncolytic viruses are promising cancer therapies due to their selective killing of tumor cells and ability to stimulate the host immune system. As an oncolytic virus platform, vaccinia virus has unique advantages, including rapid replication, a broad range of host targets, and a large capacity for transgene incorporation. In this study, we developed a novel oncolytic vaccinia virus with high potency and a favorable safety profile. We began with the International Health Department-White (IHD-W) strain, which had the strongest cytotoxicity against tumor cells among the four vaccinia virus strains tested. Next, several candidate viruses were constructed by deleting three viral genes (C11R, K3L, and J2R) in various combinations, and their efficacy and safety were compared. The virus ultimately selected, named KLS-3010, exhibited strong antitumor activity against broad targets in vitro and in vivo. Furthermore, KLS-3010 showed a favorable safety profile in mice, as determined by the biodistribution and body weight change. More promisingly, KLS-3010 was able to shift the tumor microenvironment to a proinflammatory state, as evidenced by an increase in activated lymphocytes after KLS-3010 administration, suggesting that this strain may elicit an oncolytic virus-mediated immune response. The KLS-3010 strain thus represents a promising platform for the further development of oncolytic virus-based cancer therapies.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Animales , Línea Celular Tumoral , Salud Global , Ratones , Virus Oncolíticos/genética , Distribución Tisular , Virus Vaccinia/genética , Replicación Viral
11.
Biology (Basel) ; 9(10)2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-33050357

RESUMEN

This review article focuses on the current state-of-the-art cellular and molecular biotechnology for the over-production of clinically relevant therapeutic and anabolic growth factors. We discuss how the currently available tools and emerging technologies can be used for the regenerative treatment of osteoarthritis (OA). Transfected protein packaging cell lines such as GP-293 cells may be used as "cellular factories" for large-scale production of therapeutic proteins and pro-anabolic growth factors, particularly in the context of cartilage regeneration. However, when irradiated with gamma or x-rays, these cells lose their capacity for replication, which makes them safe for use as a live cell component of intra-articular injections. This innovation is already here, in the form of TissueGene-C, a new biological drug that consists of normal allogeneic primary chondrocytes combined with transduced GP2-293 cells that overexpress the growth factor transforming growth factor ß1 (TGF-ß1). TissueGene-C has revolutionized the concept of cell therapy, allowing drug companies to develop live cells as biological drug delivery systems for direct intra-articular injection of growth factors whose half-lives are in the order of minutes. Therefore, in this paper, we discuss the potential for new innovations in regenerative medicine for degenerative diseases of synovial joints using mammalian protein production platforms, specifically protein packaging cell lines, for over-producing growth factors for cartilage tissue regeneration and give recent examples. Mammalian protein production platforms that incorporate protein packaging eukaryotic cell lines are superior to prokaryotic bacterial expression systems and are likely to have a significant impact on the development of new humanized biological growth factor therapies for treating focal cartilage defects and more generally for the treatment of degenerative joint diseases such as OA, especially when injected directly into the joint.

12.
Inflammopharmacology ; 28(5): 1237-1252, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32696209

RESUMEN

Osteoarthritis (OA) is the most common form of arthritis, characterized by cartilage destruction, pain and inflammation in the joints. Existing medications can provide relief from the symptoms, but their effects on the progression of the disease are limited. TissueGene-C (TG-C) is a novel cell and gene therapy for the treatment of OA, comprising a mixture of human allogeneic chondrocytes and irradiated cells engineered to overexpress transforming growth factor-ß1 (TGF-ß1). This study aims to investigate the efficacy and mechanism of action of TG-C in a rat model of OA. Using the monosodium-iodoacetate (MIA) model of OA, we examined whether TG-C could improve OA symptoms and cartilage structure in rats. Our results showed that TG-C provided pain relief and cartilage structural improvement in the MIA OA model over 56 days. In parallel with these long-term effects, cytokine profiles obtained on day 4 revealed increased expression of interleukin-10 (IL-10), an anti-inflammatory cytokine, in the synovial lavage fluid. Moreover, the increased levels of TGF-ß1 and IL-10 caused by TG-C induced the expression of arginase 1, a marker of M2 macrophages, and decreased the expression of CD86, a marker of M1 macrophages. These results suggest that TG-C exerts a beneficial effect on OA by inducing a M2 macrophage-dominant micro-environment. Cell therapy using TG-C may be a promising strategy for targeting the underlying pathogenic mechanisms of OA, reducing pain, improving function, and creating a pro-anabolic micro-environment. This environment supports cartilage structure regeneration and is worthy of further evaluation in future clinical trials.


Asunto(s)
Artritis Experimental/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Inflamación/terapia , Osteoartritis/terapia , Animales , Artritis Experimental/patología , Condrocitos/citología , Humanos , Inflamación/patología , Ácido Yodoacético , Macrófagos/metabolismo , Masculino , Osteoartritis/patología , Manejo del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Líquido Sinovial/metabolismo , Factor de Crecimiento Transformador beta1/genética
13.
Mol Ther Methods Clin Dev ; 18: 473-483, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32728596

RESUMEN

Neuropathic pain is a chronic pain state characterized by nerve damage, inflammation, and nociceptive neuron hyperactivity. As the underlying pathophysiology is complex, a more effective therapy for neuropathic pain would be one that targets multiple elements. Here, we generated recombinant adeno-associated viruses (AAVs) encoding three therapeutic genes, namely, glutamate decarboxylase 65, glial cell-derived neurotrophic factor, and interleukin-10, with various combinations. The efficacy for pain relief was evaluated in a rat spared nerve injury model of neuropathic pain. The maximal analgesic effect was achieved when the AAVs expressing all three genes were administered to rats with neuropathic pain. The combination of two virus constructs expressing the three genes was named KLS-2031 and evaluated as a potential novel therapeutic for neuropathic pain. Single transforaminal epidural injections of KLS-2031 into the intervertebral foramen to target the appropriate dorsal root ganglion produced notable long-term analgesic effects in female and male rats. Furthermore, KLS-2031 mitigated the neuroinflammation, neuronal cell death, and dorsal root ganglion hyperexcitability induced by the spared nerve injury. These results suggest that KLS-2031 represents a promising therapeutic option for refractory neuropathic pain.

14.
BMC Musculoskelet Disord ; 18(1): 199, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28521800

RESUMEN

BACKGROUND: Invossa™ (TissueGene-C) is a cell and gene therapy for osteoarthritis. It is composed of primary human chondrocytes (hChonJ cells) and irradiated human chondrocytes modified to express TGF-ß1 (hChonJb#7 cells). The hChonJ cells were isolated from a polydactyly donor, and TGF-ß1 cDNA was delivered to the cells, generating hChonJb#7 cells. Since the cells are allogeneic, the concern of immune response against cells has been raised. In this study, we investigated the immunogenicity of allogenic human chondrocyte, hChonJ cells. METHODS: The immunological properties of hChonJ cells were investigated through the analysis of surface marker expression and the effect on allogeneic T cell proliferation. Flow cytometry and RT-PCR analysis were performed to analyze the surface marker expression related to immune response, such as major histocompatibility complex (MHC) class I, class II, T cell co-stimulatory molecules and T cell co-inhibitory molecules. A mixed lymphocyte reaction (MLR) was conducted to evaluate how allogeneic T cells would respond to hChonJ cells. RESULTS: We observed that hChonJ cells did not express MHC class II and T cell co-stimulatory molecules, but expressed T cell co-inhibitory molecule PD-L2. IFN-γ treatment induced the expression of PD-L1, and up-regulated the expression of PD-L2. Also, we observed that hChonJ cells did not stimulate T cell proliferation from a MHC-mismatched donor. Further, they could suppress the proliferation of activated T cells. We also observed that the blockade of PD-L1 and/or PD-L2 with specific neutralizing antibody could lead to the restoration of allo-reactive T cell proliferation. CONCLUSIONS: We showed that hChonJ cells were not immunogenic but immunosuppressive, and that this phenomenon was mediated by co-inhibitory molecules PD-L1 and PD-L2 on hChonJ cells in a contact-dependent manner.


Asunto(s)
Condrocitos/inmunología , Tolerancia Inmunológica/fisiología , Inmunidad Celular/fisiología , Fenómenos Inmunogenéticos/fisiología , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo/métodos , Humanos , Inmunomodulación/fisiología
15.
Nat Commun ; 6: 8698, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26507712

RESUMEN

Induction of T-cell clonal anergy involves serial activation of transcription factors, including NFAT and Egr2/3. However, downstream effector mechanisms of these transcription factors are not fully understood yet. Here we identify Ndrg1 as an anergy factor induced by Egr2. Ndrg1 is upregulated by anergic signalling and maintained at high levels in resting anergic T cells. Overexpression of Ndrg1 mimics the anergic state and knockout of the gene prevents anergy induction. Interestingly, Ndrg1 is phosphorylated and degraded by CD28 signalling in a proteasome-dependent manner, explaining the costimulation dependence of anergy prevention. Similarly, IL-2 treatment of anergic T cells, under conditions that lead to the reversal of anergy, also induces Ndrg1 phosphorylation and degradation. Finally, older Ndrg1-deficient mice show T-cell hyperresponsiveness and Ndrg1-deficient T cells aggravate inducible autoimmune inflammation. Thus, Ndrg1 contributes to the maintenance of clonal anergy and inhibition of T-cell-mediated inflammation.


Asunto(s)
Antígenos CD28/inmunología , Proteínas de Ciclo Celular/genética , Anergia Clonal , Regulación hacia Abajo , Interleucina-2/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Linfocitos T/inmunología , Animales , Antígenos CD28/genética , Proteínas de Ciclo Celular/inmunología , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/inmunología , Interleucina-2/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
J Exp Med ; 210(4): 775-88, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23547101

RESUMEN

Airway tolerance is the usual outcome of inhalation of harmless antigens. Although T cell deletion and anergy are likely components of tolerogenic mechanisms in the lung, increasing evidence indicates that antigen-specific regulatory T cells (inducible Treg cells [iTreg cells]) that express Foxp3 are also critical. Several lung antigen-presenting cells have been suggested to contribute to tolerance, including alveolar macrophages (MØs), classical dendritic cells (DCs), and plasmacytoid DCs, but whether these possess the attributes required to directly promote the development of Foxp3(+) iTreg cells is unclear. Here, we show that lung-resident tissue MØs coexpress TGF-ß and retinal dehydrogenases (RALDH1 and RALDH 2) under steady-state conditions and that their sampling of harmless airborne antigen and presentation to antigen-specific CD4 T cells resulted in the generation of Foxp3(+) Treg cells. Treg cell induction in this model depended on both TGF-ß and retinoic acid. Transfer of the antigen-pulsed tissue MØs into the airways correspondingly prevented the development of asthmatic lung inflammation upon subsequent challenge with antigen. Moreover, exposure of lung tissue MØs to allergens suppressed their ability to generate iTreg cells coincident with blocking airway tolerance. Suppression of Treg cell generation required proteases and TLR-mediated signals. Therefore, lung-resident tissue MØs have regulatory functions, and strategies to target these cells might hold promise for prevention or treatment of allergic asthma.


Asunto(s)
Tolerancia Inmunológica , Macrófagos Alveolares/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Alérgenos/inmunología , Alérgenos/farmacología , Animales , Antineoplásicos/farmacología , Asma/genética , Asma/inmunología , Asma/patología , Asma/prevención & control , Femenino , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Linfocitos T Reguladores/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Tretinoina/farmacología
17.
Trends Immunol ; 33(3): 144-52, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22169337

RESUMEN

The tumor necrosis factor (TNF) and TNF receptor superfamilies (TNFSF and TNFRSF) consist of approximately 50 membrane and soluble proteins that can modulate cellular function. Most of these molecules are expressed by or can target cells of the immune system, and they have a wide range of actions including promoting cellular differentiation, survival, and production of inflammatory cytokines and chemokines. Emerging data show that TNFSF ligand-receptor signaling pathways are active in inflammatory and autoimmune disease. Furthermore, several genetic polymorphisms in TNFSF and TNFRSF associate with susceptibility to developing disease. Here, we examine recent data regarding the potential of these molecules as targets for therapy of autoimmune and inflammatory disease.


Asunto(s)
Inflamación/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Ligandos , Receptores del Factor de Necrosis Tumoral/inmunología
18.
J Immunol ; 187(12): 6499-507, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22079987

RESUMEN

Regulatory T cells (Tregs) play a critical role in the maintenance of airway tolerance. We report that inhaled soluble Ag induces adaptive Foxp3(+) Tregs, as well as a regulatory population of CD4(+) T cells in the lungs and lung-draining lymph nodes that express latency-associated peptide (LAP) on their cell surface but do not express Foxp3. Blocking the cytokine IL-10 or TGF-ß prevented the generation of LAP(+) Tregs and Foxp3(+) Tregs in vivo, and the LAP(+) Tregs could also be generated concomitantly with Foxp3(+) Tregs in vitro by culturing naive CD4(+) T cells with Ag and exogenous TGF-ß. The LAP(+) Tregs strongly suppressed naive CD4(+) T cell proliferation, and transfer of sorted OVA-specific LAP(+) Tregs in vivo inhibited allergic eosinophilia and Th2 cytokine expression in the lung, either when present at the time of Th2 sensitization or when injected after Th2 cells were formed. Furthermore, inflammatory innate stimuli from house dust mite extract, nucleotide-binding oligomerization domain containing 2 ligand, and LPS, which are sufficient for blocking airway tolerance, strongly decreased the induction of LAP(+) Tregs. Taken together, we concluded that inducible Ag-specific LAP(+) Tregs can suppress asthmatic lung inflammation and constitute a mediator of airway tolerance together with Foxp3(+) Tregs.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/deficiencia , Hipersensibilidad Respiratoria/patología , Hipersensibilidad Respiratoria/prevención & control , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis , Alérgenos/administración & dosificación , Alérgenos/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Epítopos de Linfocito T/biosíntesis , Epítopos de Linfocito T/genética , Factores de Transcripción Forkhead/genética , Genes Reporteros , Tolerancia Inmunológica/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/fisiología , Hipersensibilidad Respiratoria/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/genética
19.
J Immunol ; 186(12): 6972-80, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21555530

RESUMEN

TGF-ß can induce Foxp3(+) inducible regulatory T cells (Treg) and also synergize with IL-6 and IL-4 to induce Th17 and Th9 cells. We now report that NO modulates TGF-ß activity away from Treg but toward the Th1 lineage. NO potentiated Th1 differentiation in the presence of TGF-ß in both IL-12-independent and -dependent fashions by augmenting IFN-γ-activated STAT-1 and T-bet. Differentiation into Treg, Th1, and Th17 lineages could be modulated by NO competing with other cofactors, such as IL-6 and retinoic acid. NO antagonized IL-6 to block TGF-ß-directed Th17 differentiation, and together with IL-6, NO suppressed Treg development induced by TGF-ß and retinoic acid. Furthermore, we show that physiologically produced NO from TNF and inducible NO synthase-producing dendritic cells can contribute to Th1 development predominating over Treg development through a synergistic activity induced when these cells cocluster with conventional dendritic cells presenting Ag to naive Th cells. This illustrates that NO is another cofactor allowing TGF-ß to participate in development of multiple Th lineages and suggests a new mechanism by which NO, which is associated with protection against intracellular pathogens, might maintain effective Th1 immunity.


Asunto(s)
Diferenciación Celular/inmunología , Óxido Nítrico/farmacología , Linfocitos T Reguladores/citología , Células TH1/citología , Factor de Crecimiento Transformador beta/inmunología , Animales , Presentación de Antígeno , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/inmunología , Ratones , Óxido Nítrico/inmunología , Transducción de Señal/inmunología , Células TH1/efectos de los fármacos
20.
Nat Med ; 17(5): 596-603, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21499267

RESUMEN

Individuals with chronic asthma show a progressive decline in lung function that is thought to be due to structural remodeling of the airways characterized by subepithelial fibrosis and smooth muscle hyperplasia. Here we show that the tumor necrosis factor (TNF) family member LIGHT is expressed on lung inflammatory cells after allergen exposure. Pharmacological inhibition of LIGHT using a fusion protein between the IgG Fc domain and lymphotoxin ß receptor (LTßR) reduces lung fibrosis, smooth muscle hyperplasia and airway hyperresponsiveness in mouse models of chronic asthma, despite having little effect on airway eosinophilia. LIGHT-deficient mice also show a similar impairment in fibrosis and smooth muscle accumulation. Blockade of LIGHT suppresses expression of lung transforming growth factor-ß (TGF-ß) and interleukin-13 (IL-13), cytokines implicated in remodeling in humans, whereas exogenous administration of LIGHT to the airways induces fibrosis and smooth muscle hyperplasia, Thus, LIGHT may be targeted to prevent asthma-related airway remodeling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Asma/etiología , Asma/patología , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/fisiología , Interleucina-13/fisiología , Pulmón/patología , Pulmón/fisiopatología , Heterotrímero de Linfotoxina alfa1 y beta2/antagonistas & inhibidores , Heterotrímero de Linfotoxina alfa1 y beta2/fisiología , Ratones , Ratones Noqueados , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA