Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Commun ; 14(1): 5666, 2023 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-37723181

RESUMO

TANK-binding kinase 1 (TBK1) is a key signalling component in the production of type-I interferons, which have essential antiviral activities, including against SARS-CoV-2. TBK1, and its homologue IκB kinase-ε (IKKε), can also induce pro-inflammatory responses that contribute to pathogen clearance. While initially protective, sustained engagement of type-I interferons is associated with damaging hyper-inflammation found in severe COVID-19 patients. The contribution of TBK1/IKKε signalling to these responses is unknown. Here we find that the small molecule idronoxil inhibits TBK1/IKKε signalling through destabilisation of TBK1/IKKε protein complexes. Treatment with idronoxil, or the small molecule inhibitor MRT67307, suppresses TBK1/IKKε signalling and attenuates cellular and molecular lung inflammation in SARS-CoV-2-challenged mice. Our findings additionally demonstrate that engagement of STING is not the major driver of these inflammatory responses and establish a critical role for TBK1/IKKε signalling in SARS-CoV-2 hyper-inflammation.


Assuntos
COVID-19 , Interferon Tipo I , Animais , Camundongos , Quinase I-kappa B , Modelos Animais de Doenças , SARS-CoV-2 , Inflamação
2.
mBio ; 13(4): e0206422, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35924852

RESUMO

Cytoplasmic detection of DNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) is an essential component of antiviral responses. Upon synthesis, cGAMP binds to the stimulator of interferon (IFN) genes (STING) in infected and adjacent cells through intercellular transfer by connexins forming gap-junctions, eliciting a strong IFN-ß-driven antiviral response. We demonstrate here that Genistein, a flavonoid compound naturally occurring in soy-based foods, inhibits cGAS-STING antiviral signaling at two levels. First, Genistein pretreatment of cGAMP-producing cells inhibited gap-junction intercellular communication, resulting in reduced STING responses in adjacent cells. In addition, Genistein directly blocked STING activation by the murine agonist DMXAA, by decreasing the interaction of STING with TBK1 and IKKε. As a result, Genistein attenuated STING signaling in human and mouse cells, dampening antiviral activity against Semliki Forest Virus infection. Collectively, our findings identify a previously unrecognized proviral activity of Genistein mediated via its inhibitory effects at two levels of cGAS-STING signaling. IMPORTANCE Several reports suggest that Genistein exhibits antiviral activities against DNA viruses. Our work uncovers a previously unrecognized proviral effect of Genistein, through inhibition of the cGAS-STING pathway at the level of cGAMP transfer and its sensing by STING. This suggests that the use of Genistein as an antiviral should be taken with caution as it may reduce the protective antiviral effects elicited by host STING activation.


Assuntos
Genisteína , Proteínas de Membrana , Animais , Antivirais/farmacologia , Genisteína/farmacologia , Humanos , Imunidade Inata/genética , Interferon beta/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/genética
3.
Am J Respir Cell Mol Biol ; 62(4): 413-422, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31967851

RESUMO

Pirfenidone (PFD) is a pharmacological compound with therapeutic efficacy in idiopathic pulmonary fibrosis. It has been chiefly characterized as an antifibrotic agent, although it was initially developed as an antiinflammatory compound because of its ability to diminish the accumulation of inflammatory cells and cytokines. Despite recent studies that have elucidated key mechanisms, the precise molecular activities of PFD remain incompletely understood. PFD modulates fibrogenic growth factors, thereby attenuating fibroblast proliferation, myofibroblast differentiation, collagen and fibronectin synthesis, and deposition of extracellular matrix. This effect is mediated by suppression of TGF-ß1 (transforming growth factor-ß1) and other growth factors. Here, we appraise the impact of PFD on TGF-ß1 production and its downstream pathways. Accumulating evidence indicates that PFD also downregulates inflammatory pathways and therefore has considerable potential as a viable and innovative antiinflammatory compound. We examine the effects of PFD on inflammatory cells and the production of pro- and antiinflammatory cytokines in the lung. In this context, recent evidence that PFD can target inflammasome pathways and ensuing lung inflammation is highlighted. Finally, the antioxidant properties of PFD, such as its ability to inhibit redox reactions and regulate oxidative stress-related genes and enzymes, are detailed. In summary, this narrative review examines molecular mechanisms underpinning PFD and its recognized benefits in lung fibrosis. We highlight preclinical data that demonstrate the potential of PFD as a nonsteroidal antiinflammatory agent and outline areas for future research.


Assuntos
Pneumopatias/tratamento farmacológico , Piridonas/farmacologia , Piridonas/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Humanos , Pulmão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
4.
mBio ; 8(5)2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974621

RESUMO

Inflammatory responses, while essential for pathogen clearance, can also be deleterious to the host. Chemical inhibition of topoisomerase 1 (Top1) by low-dose camptothecin (CPT) can suppress transcriptional induction of antiviral and inflammatory genes and protect animals from excessive and damaging inflammatory responses. We describe the unexpected finding that minor DNA damage from topoisomerase 1 inhibition with low-dose CPT can trigger a strong antiviral immune response through cyclic GMP-AMP synthase (cGAS) detection of cytoplasmic DNA. This argues against CPT having only anti-inflammatory activity. Furthermore, expression of the simian virus 40 (SV40) large T antigen was paramount to the proinflammatory antiviral activity of CPT, as it potentiated cytoplasmic DNA leakage and subsequent cGAS recruitment in human and mouse cell lines. This work suggests that the capacity of Top1 inhibitors to blunt inflammatory responses can be counteracted by viral oncogenes and that this should be taken into account for their therapeutic development.IMPORTANCE Recent studies suggest that low-dose DNA-damaging compounds traditionally used in cancer therapy can have opposite effects on antiviral responses, either suppressing (with the example of CPT) or potentiating (with the example of doxorubicin) them. Our work demonstrates that the minor DNA damage promoted by low-dose CPT can also trigger strong antiviral responses, dependent on the presence of viral oncogenes. Taken together, these results call for caution in the therapeutic use of low-dose chemotherapy agents to modulate antiviral responses in humans.


Assuntos
DNA Topoisomerases Tipo I/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Nucleotídeos Cíclicos/metabolismo , Vírus 40 dos Símios/efeitos dos fármacos , Inibidores da Topoisomerase I/farmacologia , Animais , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/imunologia , Antivirais/farmacologia , Camptotecina/farmacologia , Linhagem Celular , Técnicas de Cocultura , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , Inflamação , Camundongos , Vírus 40 dos Símios/imunologia , Vírus 40 dos Símios/fisiologia , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/virologia
5.
Clin Sci (Lond) ; 131(14): 1713-1721, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28667069

RESUMO

Since its discovery in 2001, human metapneumovirus (hMPV) has been identified as an important cause of respiratory tract infection in young children, second only to the closely related respiratory syncytial virus (RSV). Clinical evidence suggests that hMPV is associated with acute exacerbations of asthma in both children and adults, and may play a role in initiating asthma development in children. Animal models have demonstrated that airway hyperresponsiveness (AHR) and inflammation are triggered following hMPV infection, and hMPV is able to persist in vivo by inhibiting innate immune responses and causing aberrant adaptive responses. In this review, we discuss the prevalence of hMPV infection in pediatric and adult populations and its potential role in asthma exacerbation. We also review recent advances made in animal models to determine immune responses following hMPV infection, and compare to what is known about RSV.


Assuntos
Asma/virologia , Metapneumovirus , Infecções por Paramyxoviridae/complicações , Infecções por Vírus Respiratório Sincicial/complicações , Vírus Sincicial Respiratório Humano , Doença Aguda , Animais , Modelos Animais de Doenças , Humanos , Imunidade Inata , Infecções por Paramyxoviridae/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia
6.
J Infect Dis ; 215(10): 1536-1545, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28379462

RESUMO

Background: Human metapneumovirus (hMPV) infection is implicated in exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Research into the pathogenesis of infection is restricted to animal models, and information about hMPV replication and inflammatory and immune responses in human disease is limited. Methods: Human primary bronchial epithelial cells (PBECs) from healthy and asthmatic subjects and those with COPD were infected with hMPV, with or without glucocorticosteroid (GCS) exposure. Viral replication, inflammatory and immune responses, and apoptosis were analyzed. We also determined whether adjuvant interferon (IFN) can blunt hMPV infection in vitro and in a murine model. Results: hMPV infected human PBECs and viral replication was enhanced in cells from patients with COPD. The virus induced gene expression of IFN-stimulated gene 56 (ISG56) and IFN-ß, as well as IFN-γ-inducible protein 10 (IP-10) and regulated on activation, normal T cell expressed and secreted (RANTES), and more so in cells from patients with COPD. GCS exposure enhanced hMPV replication despite increased IFN expression. Augmented virus replication associated with GCS was mediated by reduced apoptosis via induction of antiapoptotic genes. Adjuvant IFN treatment suppressed hMPV replication in PBECs and reduced hMPV viral titers and inflammation in vivo. Conclusions: hMPV infects human PBECs, eliciting innate and inflammatory responses. Replication is enhanced by GCS and adjuvant IFN is an effective treatment, restricting virus replication and proinflammatory consequences of hMPV infections.


Assuntos
Glucocorticoides/farmacologia , Interferon gama/farmacologia , Metapneumovirus , Infecções por Paramyxoviridae/virologia , Doença Pulmonar Obstrutiva Crônica/virologia , Animais , Apoptose/efeitos dos fármacos , Asma/virologia , Brônquios/citologia , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Mucosa Respiratória/citologia , Replicação Viral/efeitos dos fármacos
7.
Nucleic Acids Res ; 45(1): 198-205, 2017 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-27694309

RESUMO

Acridine dyes, including proflavine and acriflavine, were commonly used as antiseptics before the advent of penicillins in the mid-1940s. While their mode of action on pathogens was originally attributed to their DNA intercalating activity, work in the early 1970s suggested involvement of the host immune responses, characterized by induction of interferon (IFN)-like activities through an unknown mechanism. We demonstrate here that sub-toxic concentrations of a mixture of acriflavine and proflavine instigate a cyclic-GMP-AMP (cGAMP) synthase (cGAS)-dependent type-I IFN antiviral response. This pertains to the capacity of these compounds to induce low level DNA damage and cytoplasmic DNA leakage, resulting in cGAS-dependent cGAMP-like activity. Critically, acriflavine:proflavine pre-treatment of human primary bronchial epithelial cells significantly reduced rhinovirus infection. Collectively, our findings constitute the first evidence that non-toxic DNA binding agents have the capacity to act as indirect agonists of cGAS, to exert potent antiviral effects in mammalian cells.


Assuntos
Acriflavina/farmacologia , Antivirais/farmacologia , Fatores Imunológicos/farmacologia , Substâncias Intercalantes/farmacologia , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , Proflavina/farmacologia , Animais , Brônquios/efeitos dos fármacos , Brônquios/imunologia , Brônquios/virologia , Linhagem Celular Transformada , Chlorocebus aethiops , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Proteínas de Membrana/agonistas , Proteínas de Membrana/imunologia , Camundongos , Nucleotídeos Cíclicos/imunologia , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/imunologia , Cultura Primária de Células , Rhinovirus/efeitos dos fármacos , Rhinovirus/crescimento & desenvolvimento , Transdução de Sinais , Células Vero , Carga Viral/efeitos dos fármacos
8.
Am J Respir Cell Mol Biol ; 55(6): 759-766, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27603223

RESUMO

Transforming growth factor-ß (TGFB) regulates cell proliferation, differentiation, apoptosis, and matrix homeostasis and is intimately involved in fibrosis. TGFB expression is increased in fibrotic lung diseases, such as idiopathic pulmonary fibrosis, and in chronic inflammatory conditions, such as chronic obstructive pulmonary disease and asthma. In addition to exhibiting profibrotic activities, the protein exhibits profound immune-suppressive actions involving both innate and adaptive responses, but often this aspect of TGFB biology is overlooked. Recent investigations have demonstrated that TGFB causes wide-ranging immune suppression, including blunting of pivotal early innate IFN responses. These activities permit severe virus infections, often followed by secondary bacterial infections, which may last longer, with augmented inflammation, scarring, fibrosis, and loss of lung function. Strategies to oppose TGFB actions or to enhance IFN responses may help ameliorate the detrimental consequences of infection in patients with diseases characterized by TGFB overexpression, inflammation, and fibrosis.


Assuntos
Imunidade Inata , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Pulmão/virologia , Modelos Biológicos , Fibrose Pulmonar/microbiologia , Fibrose Pulmonar/virologia , Transdução de Sinais
9.
Sci Rep ; 4: 7176, 2014 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-25417801

RESUMO

Glucocorticosteroids (GCS) are used on a daily basis to reduce airway inflammation in asthma and chronic obstructive pulmonary disease (COPD). This treatment is usually escalated during acute disease exacerbations, events often associated with virus infections. We examined the impact of GCS on anti-viral defences and virus replication and assessed supplementary interferon (IFN) treatment. Here, we report that treatment of primary human airway cells in vitro with GCS prior to rhinovirus (RV) or influenza A virus (IAV) infection significantly reduces the expression of innate anti-viral genes and increases viral replication. Mice given intranasal treatment with GCS prior to IAV infection developed more severe disease associated with amplified virus replication and elevated inflammation in the airways. Adjuvant IFN treatment markedly reduced GCS-amplified infections in human airway cells and in mouse lung. This study demonstrates that GCS cause an extrinsic compromise in anti-viral defences, enhancing respiratory virus infections and provides a rationale for adjuvant IFN treatment.


Assuntos
Glucocorticoides/farmacologia , Vírus da Influenza A/fisiologia , Interferons/farmacologia , Rhinovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Animais , Antivirais/farmacologia , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/virologia , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/veterinária , Índice de Gravidade de Doença
10.
Cell Host Microbe ; 15(5): 623-35, 2014 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-24746552

RESUMO

The intracellular innate immune receptor NOD1 detects Gram-negative bacterial peptidoglycan (PG) to induce autophagy and inflammatory responses in host cells. To date, the intracellular compartment in which PG is detected by NOD1 and whether NOD1 directly interacts with PG are two questions that remain to be resolved. To address this, we used outer membrane vesicles (OMVs) from pathogenic bacteria as a physiological mechanism to deliver PG into the host cell cytosol. We report that OMVs induced autophagosome formation and inflammatory IL-8 responses in epithelial cells in a NOD1- and RIP2-dependent manner. PG contained within OMVs colocalized with both NOD1 and RIP2 in EEA1-positive early endosomes. Further, we provide evidence for direct interactions between NOD1 and PG. Collectively, these findings demonstrate that NOD1 detects PG within early endosomes, thereby promoting RIP2-dependent autophagy and inflammatory signaling in response to bacterial infection.


Assuntos
Autofagia , Endossomos/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Peptidoglicano/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/imunologia , Receptores Imunológicos/imunologia , Animais , Linhagem Celular , Endossomos/microbiologia , Infecções por Helicobacter/enzimologia , Infecções por Helicobacter/genética , Helicobacter pylori/fisiologia , Humanos , Camundongos , Proteína Adaptadora de Sinalização NOD1/genética , Ligação Proteica , Infecções por Pseudomonas/enzimologia , Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/fisiologia , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Receptores Imunológicos/genética , Transdução de Sinais
11.
PLoS One ; 8(8): e71316, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951130

RESUMO

Human Rhinovirus (HRV) infection results in shut down of essential cellular processes, in part through disruption of nucleocytoplasmic transport by cleavage of the nucleoporin proteins (Nups) that make up the host cell nuclear pore. Although the HRV genome encodes two proteases (2A and 3C) able to cleave host proteins such as Nup62, little is known regarding the specific contribution of each. Here we use transfected as well as HRV-infected cells to establish for the first time that 3C protease is most likely the mediator of cleavage of Nup153 during HRV infection, while Nup62 and Nup98 are likely to be targets of HRV2A protease. HRV16 3C protease was also able to elicit changes in the appearance and distribution of the nuclear speckle protein SC35 in transfected cells, implicating it as a key mediator of the mislocalisation of SC35 in HRV16-infected cells. In addition, 3C protease activity led to the redistribution of the nucleolin protein out of the nucleolus, but did not affect nuclear localisation of hnRNP proteins, implying that complete disruption of nucleocytoplasmic transport leading to relocalisation of hnRNP proteins from the nucleus to the cytoplasm in HRV-infected cells almost certainly requires 2A in addition to 3C protease. Thus, a specific role for HRV 3C protease in cleavage and mislocalisation of host cell nuclear proteins, in concert with 2A, is implicated for the first time in HRV pathogenesis.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Transporte Ativo do Núcleo Celular , Animais , Western Blotting , Células COS , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Cisteína Endopeptidases/genética , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Microscopia Confocal , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteólise , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Rhinovirus/genética , Rhinovirus/metabolismo , Rhinovirus/fisiologia , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Transfecção , Proteínas Virais/genética , Nucleolina
12.
Immunol Cell Biol ; 91(4): 321-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23399697

RESUMO

The innate immune response to virus must be balanced to eliminate infection yet limit damaging inflammation. A critical arm of the antiviral response is launched by the retinoic acid-inducible-gene I (RIG-I) protein. RIG-I is activated by viral RNA then associates with the mitochondrial antiviral signaling (MAVS) protein to subsequently induce potent inflammatory cytokines. Here, we demonstrate the mitochondrial E3 ubiquitin protein ligase 1 (MUL1) is a crucial moderator of RIG-I signaling. MUL1 is localized to the mitochondria where it interacts with MAVS and catalyzes RIG-I post-translational modifications that inhibit RIG-I-dependent cell signaling. Accordingly, depletion of MUL1 potentiated RIG-I mediated nuclear factor-kappa B (NF-κB) and interferon (IFN) ß reporter activity. Moreover, depletion of MUL1 boosted the antiviral response and increased proinflammatory cytokines following challenge with the RNA mimetic poly I:C and Sendai virus. We therefore submit that MUL1 is a novel regulator of the RIG-I-like receptor-dependent antiviral response, that otherwise functions to limit inflammation.


Assuntos
Antivirais/metabolismo , Mitocôndrias/metabolismo , Transdução de Sinais/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quimiocina CCL5/metabolismo , Citocinas/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Células HEK293 , Humanos , Inflamação/patologia , Poliubiquitina/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transporte Proteico , Receptores Imunológicos , Proteína SUMO-1/metabolismo , Ubiquitinação
13.
Am J Respir Cell Mol Biol ; 41(3): 339-47, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19168696

RESUMO

Individuals with asthma are prone to viral and bacterial infections, and most asthma exacerbations have been linked to viruses, particularly rhinovirus. Excess transforming growth factor (TGF)-beta present in asthmatic airways may cause immune suppression, as well as transdifferentiate fibroblasts to myofibroblasts, thereby augmenting proinflammatory responses after rhinovirus infection. After rhinovirus infection we examined virus replication and host cell immune responses in airway fibroblasts in the presence of TGF-beta1 and in myofibroblasts. Primary culture fibroblasts were pretreated with TGF-beta1 or transdifferentiated into myofibroblasts, and then infected with rhinovirus. Viral replication, virus release, chemokine production, and interferon (IFN) responses were measured over 72 hours. Rhinovirus replication and virus release into supernatants were enhanced in fibroblasts incubated with TGF-beta1 and in fibroblasts obtained from patients with asthma. Myofibroblasts also showed more rhinovirus replication, and infected myofibroblasts produced excess neutrophil chemokines. Examination of innate responses revealed blunting of type I IFN reactions with dissociated viral RNA and IFN mRNA responses. Addition of type I IFN restituted antiviral responses, and the effect of TGF-beta1 appeared to be mediated via actions on IFN regulatory factor-3 pathways. These data demonstrate that TGF-beta1 mediates enhanced virus replication and proinflammatory responses in airway cells. TGF-beta may act as an endogenous immunosuppressant promoting virus replication and inflammation during the evolution of acute severe asthma associated with rhinovirus infection.


Assuntos
Imunidade Inata/fisiologia , Infecções por Picornaviridae/imunologia , Rhinovirus/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Asma/imunologia , Asma/virologia , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Quimiocinas/imunologia , Criança , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/virologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Interleucina-8/genética , Interleucina-8/metabolismo , Neutrófilos/imunologia , Mucosa Respiratória/citologia , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia
14.
Pediatr Res ; 60(3): 309-14, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16857766

RESUMO

Reduction of an enzyme activity required for the lysosomal degradation of glycosaminoglycan (gag) chains will result in a mucopolysaccharidosis (MPS) disorder. Substrate deprivation therapy (SDT), a potential therapy option for MPS with residual enzyme activity, aims to reduce the synthesis of gag chains, the natural substrate for the deficient enzyme. Reduced substrate levels would balance the reduced level of enzyme in patient cells, resulting in normalized gag turnover. Rhodamine B, a nonspecific inhibitor, reduced gag synthesis in a range of normal and MPS cells and also decreased lysosomal storage of gag in MPS VI (72%) and MPS IIIA (60%) cells. Body weight gain of male MPS IIIA mice treated with 1 mg/kg rhodamine B was reduced compared with untreated MPS IIIA mice and was indistinguishable from that of normal mice. Liver size, total gag content, and lysosomal gag was reduced in treated MPS IIIA animals as was urinary gag excretion. Lysosomal gag content in the brain was also reduced by treatment. The alteration in MPS IIIA clinical pathology by rhodamine B, combined with the observation that treatment had no effect on the health of normal animals, demonstrates the potential for SDT in general as a therapy for MPS disorders.


Assuntos
Glicosaminoglicanos/antagonistas & inibidores , Glicosaminoglicanos/biossíntese , Mucopolissacaridose III/tratamento farmacológico , Mucopolissacaridose III/metabolismo , Rodaminas/uso terapêutico , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Glicosaminoglicanos/urina , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Mucopolissacaridose III/patologia , Tamanho do Órgão/efeitos dos fármacos , Aumento de Peso/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA