Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Toxicol Pathol ; 51(5): 246-256, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37921115

RESUMO

Replication-incompetent adeno-associated virus (AAV)-based vectors are nonpathogenic viral particles used to deliver therapeutic genes to treat multiple monogenic disorders. AAVs can elicit immune responses; thus, one challenge in AAV-based gene therapy is the presence of neutralizing antibodies against vector capsids that may prevent transduction of target cells or elicit adverse findings. We present safety findings from two 12-week studies in nonhuman primates (NHPs) with pre-existing or treatment-emergent antibodies. In the first study, NHPs with varying levels of naturally acquired anti-AAV5 antibodies were dosed with an AAV5-based vector encoding human factor VIII (hFVIII). In the second study, NHPs with no pre-existing anti-AAV antibodies were dosed with an AAV5-based vector carrying the beta subunit of choriogonadotropic hormone (bCG); this led to the induction of high-titer antibodies against the AAV5 capsid. Four weeks later, the same NHPs received an equivalent dose of an AAV5-based vector carrying human factor IX (hFIX). In both of these studies, the administration of vectors carrying hFVIII, bCG, and hFIX was well-tolerated in NHPs with no adverse clinical pathology or microscopic findings. These two studies demonstrate the safety of AAV-based vector administration in NHPs with either low-titer pre-existing anti-AAV5 antibodies or re-administration, even in the presence of high-titer antibodies.


Assuntos
Capsídeo , Dependovirus , Animais , Humanos , Dependovirus/genética , Anticorpos Neutralizantes/genética , Terapia Genética
2.
Toxicol Rep ; 10: 357-366, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36923444

RESUMO

Mucopolysaccharidosis Type IIIB (MPS IIIB) is an ultrarare, fatal pediatric disease with no approved therapy. It is caused by mutations in the gene encoding for lysosomal enzyme alpha-N-acetylglucosaminidase (NAGLU). Tralesinidase alfa (TA) is a fusion protein comprised of recombinant NAGLU and a modified human insulin-like growth factor 2 that is being developed as an enzyme replacement therapy for MPS IIIB. Since MPS IIIB is a pediatric disease the safety/toxicity, pharmacokinetics and biodistribution of TA were evaluated in juvenile non-human primates that were administered up to 5 weekly intracerebroventricular (ICV) or single intravenous (IV) infusions of TA. TA administered by ICV slow-, ICV isovolumetric bolus- or IV-infusion was well-tolerated, and no effects were observed on clinical observations, electrocardiographic or ophthalmologic parameters, or respiratory rates. The drug-related changes observed were limited to increased cell infiltrates in the CSF and along the ICV catheter track after ICV administration. These findings were not associated with functional changes and are associated with the use of ICV catheters. The CSF PK profiles were consistent across all conditions tested and TA distributed widely in the CNS after ICV administration. Anti-drug antibodies were observed but did not appear to significantly affect the exposure to TA. Correlations between TA concentrations in plasma and brain regions in direct contact with the cisterna magna suggest glymphatic drainage may be responsible for clearance of TA from the CNS. The data support the administration of TA by isovolumetric bolus ICV infusion to pediatric patients with MPS IIIB.

3.
J Pharmacol Exp Ther ; 382(3): 277-286, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35717448

RESUMO

Mucopolysaccharidosis type IIIB (MPS IIIB; Sanfilippo syndrome B; OMIM #252920) is a lethal, pediatric, neuropathic, autosomal recessive, and lysosomal storage disease with no approved therapy. Patients are deficient in the activity of N-acetyl-alpha-glucosaminidase (NAGLU; EC 3.2.150), necessary for normal lysosomal degradation of the glycosaminoglycan heparan sulfate (HS). Tralesinidase alfa (TA), a fusion protein comprised of recombinant human NAGLU and a modified human insulin-like growth factor 2, is in development as an enzyme replacement therapy that is administered via intracerebroventricular (ICV) infusion, thus circumventing the blood brain barrier. Previous studies have confirmed ICV infusion results in widespread distribution of TA throughout the brains of mice and nonhuman primates. We assessed the long-term tolerability, pharmacology, and clinical efficacy of TA in a canine model of MPS IIIB over a 20-month study. Long-term administration of TA was well tolerated as compared with administration of vehicle. TA was widely distributed across brain regions, which was confirmed in a follow-up 8-week pharmacokinetic/pharmacodynamic study. MPS IIIB dogs treated for up to 20 months had near-normal levels of HS and nonreducing ends of HS in cerebrospinal fluid and central nervous system (CNS) tissues. TA-treated MPS IIIB dogs performed better on cognitive tests and had improved CNS pathology and decreased cerebellar volume loss relative to vehicle-treated MPS IIIB dogs. These findings demonstrate the ability of TA to prevent or limit the biochemical, pathologic, and cognitive manifestations of canine MPS IIIB disease, thus providing support of its potential long-term tolerability and efficacy in MPS IIIB subjects. SIGNIFICANCE STATEMENT: This work illustrates the efficacy and tolerability of tralesinidase alfa as a potential therapeutic for patients with mucopolysaccharidosis type IIIB (MPS IIIB) by documenting that administration to the central nervous system of MPS IIIB dogs prevents the accumulation of disease-associated glycosaminoglycans in lysosomes, hepatomegaly, cerebellar atrophy, and cognitive decline.


Assuntos
Mucopolissacaridose III , Animais , Encéfalo/metabolismo , Criança , Modelos Animais de Doenças , Cães , Terapia de Reposição de Enzimas , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/líquido cefalorraquidiano , Heparitina Sulfato/uso terapêutico , Humanos , Mucopolissacaridose III/tratamento farmacológico , Mucopolissacaridose III/patologia
4.
Drug Deliv Transl Res ; 10(2): 425-439, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31942701

RESUMO

BMN 250 is being developed as enzyme replacement therapy for Sanfilippo type B, a primarily neurological rare disease, in which patients have deficient lysosomal alpha-N-acetylglucosaminidase (NAGLU) enzyme activity. BMN 250 is taken up in target cells by the cation-independent mannose 6-phosphate receptor (CI-MPR, insulin-like growth factor 2 receptor), which then facilitates transit to the lysosome. BMN 250 is dosed directly into the central nervous system via the intracerebroventricular (ICV) route, and the objective of this work was to compare systemic intravenous (IV) and ICV delivery of BMN 250 to confirm the value of ICV dosing. We first assess the ability of enzyme to cross a potentially compromised blood-brain barrier in the Naglu-/- mouse model and then assess the potential for CI-MPR to be employed for receptor-mediated transport across the blood-brain barrier. In wild-type and Naglu-/- mice, CI-MPR expression in brain vasculature is high during the neonatal period but virtually absent by adolescence. In contrast, CI-MPR remains expressed through adolescence in non-affected non-human primate and human brain vasculature. Combined results from IV administration of BMN 250 in Naglu-/- mice and IV and ICV administration in healthy juvenile non-human primates suggest a limitation to therapeutic benefit from IV administration because enzyme distribution is restricted to brain vascular endothelial cells: enzyme does not reach target neuronal cells following IV administration, and pharmacological response following IV administration is likely restricted to clearance of substrate in endothelial cells. In contrast, ICV administration enables central nervous system enzyme replacement with biodistribution to target cells.


Assuntos
Acetilglucosaminidase/administração & dosagem , Acetilglucosaminidase/genética , Barreira Hematoencefálica/química , Fator de Crescimento Insulin-Like II/administração & dosagem , Mucopolissacaridose III/tratamento farmacológico , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Acetilglucosaminidase/uso terapêutico , Administração Intravenosa , Animais , Modelos Animais de Doenças , Terapia de Reposição de Enzimas , Feminino , Infusões Intraventriculares , Fator de Crescimento Insulin-Like II/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Mucopolissacaridose III/genética , Primatas , Proteínas Recombinantes de Fusão/uso terapêutico , Pesquisa Translacional Biomédica
5.
Ann Clin Transl Neurol ; 6(12): 2437-2447, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31814335

RESUMO

OBJECTIVE: Neuronal ceroid lipofuscinosis type 2 (CLN2 disease) is a rare, progressive, fatal neurodegenerative pediatric disorder resulting from deficiencies of the lysosomal enzyme tripeptidyl peptidase 1 that are caused by mutations in TPP1. Identifying biomarkers of CLN2 disease progression will be important in assessing the efficacy of therapeutic interventions for this disorder. Neurofilament light is an intrinsic component of healthy neurons; elevated circulating extracellular neurofilament light is a biomarker of neuropathology in several adult-onset neurological diseases. Our objective was to assess whether circulating neurofilament light is a biomarker that is responsive to enzyme replacement therapy (ERT) in CLN2 disease. METHODS: Using an ultrasensitive immunoassay, we assessed plasma neurofilament light changes during disease progression in a canine model of CLN2 disease and in ERT clinical trial CLN2 disease patients. RESULTS: In tripeptidyl peptidase 1 (TPP1)-null dogs (N = 11), but not in control dogs [N = 6 (TPP1+/- ) and N = 27 (WT)], neurofilament light levels increased more than tenfold above initial low baseline levels during disease progression. Before treatment in 21 human subjects with CLN2 disease (age range: 1.72-6.85 years), neurofilament light levels were 48-fold higher (P < 0.001) than in 7 pediatric controls (age range: 8-11 years). Pretreatment neurofilament light did not significantly correlate with disease severity or age. In CLN2 disease subjects receiving ERT, neurofilament light levels decreased by 50% each year over more than 3 years of treatment. INTERPRETATION: Our data indicate that circulating neurofilament light is a treatment-responsive biomarker in CLN2 disease and could contribute to understanding of the pathophysiology of this devastating pediatric disorder.


Assuntos
Aminopeptidases/farmacologia , Dipeptidil Peptidases e Tripeptidil Peptidases/farmacologia , Progressão da Doença , Terapia de Reposição de Enzimas , Proteínas de Neurofilamentos/sangue , Lipofuscinoses Ceroides Neuronais/sangue , Serina Proteases/farmacologia , Aminopeptidases/genética , Animais , Animais Geneticamente Modificados , Biomarcadores/sangue , Criança , Pré-Escolar , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Modelos Animais de Doenças , Cães , Feminino , Humanos , Lactente , Masculino , Proteínas de Neurofilamentos/efeitos dos fármacos , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Serina Proteases/genética , Tripeptidil-Peptidase 1
6.
J Vis Exp ; (139)2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30247460

RESUMO

The administration of enzyme replacement therapies (ERTs) and other biologic therapies to patients may elicit an anti-drug immune response. The characterization of these anti-drug antibodies (ADA), especially those that may neutralize the biological activity of the drug, termed neutralizing antibodies (NAbs), is crucial in understanding the effects of these antibodies on the drug's pharmacological profile. This protocol describes a cell-based flow cytometry method to detect factors that neutralize the cellular uptake of a representative lysosomal ERT in human matrix. The protocol consists of three procedures: screening, a confirmatory step, and titer assays to detect, identify, and establish the relative level of neutralizing antibody titer in subject samples. In this method, samples are first mixed with the fluorophore-conjugated ERT product, then incubated with cells [e.g., human T lymphocytes (Jurkat cells)] that express a cell-surface cation-independent mannose 6-phosphate receptor (CI-M6PR), and finally, analyzed with a flow cytometer. A sample without NAbs will result in the uptake of the fluorophore-conjugated ERT product via CI-M6PR, whereas, the presence of NAbs will bind to the drug and interfere with the CI-M6PR binding and uptake. The amount of the fluorophore-conjugated ERT internalized by the Jurkat cells is measured by flow cytometry and evaluated as the percentage (%) signal inhibition compared to the response obtained in the presence of a representative drug-naïve matrix. In the confirmatory step, the samples are pre-incubated with ERT-conjugated magnetic beads to deplete drug-specific factors that bind to the drug (such as NAbs) prior to an incubation with cells. Samples that screen and confirm positive for drug-specific NAbs in the assay are then serially diluted to generate an antibody titer. Semi-quantitative antibody titers may be correlated with measurements of drug safety and efficacy.


Assuntos
Anticorpos Neutralizantes/imunologia , Bioensaio/métodos , Terapia de Reposição de Enzimas , Anticorpos Neutralizantes/metabolismo , Transporte Biológico , Humanos , Células Jurkat
7.
J Immunol Methods ; 440: 41-51, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27789297

RESUMO

Many enzyme replacement therapies (ERTs) for lysosomal storage disorders use the cell-surface cation-independent mannose-6 phosphate receptor (CI-M6PR) to deliver ERTs to the lysosome. However, neutralizing antibodies (NAb) may interfere with this process. We previously reported that most individuals with Morquio A who received elosulfase alfa in the phase 3 MOR-004 trial tested positive for NAbs capable of interfering with binding to CI-M6PR ectodomain in an ELISA-based assay. However, no correlation was detected between NAb occurrence and clinical efficacy or pharmacodynamics. To quantify and better characterize the impact of NAbs, we developed a functional cell-based flow cytometry assay with a titer step that detects antibodies capable of interfering with elosulfase alfa uptake. Serum samples collected during the MOR-004 trial were tested and titers were determined. Consistent with earlier findings on NAb positivity, no correlations were observed between NAb titers and the clinical outcomes of elosulfase alfa-treated individuals with Morquio A.


Assuntos
Anticorpos Neutralizantes/sangue , Condroitina Sulfatases/uso terapêutico , Terapia de Reposição de Enzimas/métodos , Citometria de Fluxo , Mucopolissacaridose IV/tratamento farmacológico , Receptor IGF Tipo 2/imunologia , Testes Sorológicos/métodos , Anticorpos Neutralizantes/imunologia , Transporte Biológico , Condroitina Sulfatases/farmacocinética , Método Duplo-Cego , Humanos , Células Jurkat , Microscopia Confocal , Mucopolissacaridose IV/sangue , Mucopolissacaridose IV/enzimologia , Mucopolissacaridose IV/imunologia , Receptor IGF Tipo 2/metabolismo , Fatores de Tempo , Resultado do Tratamento
8.
Dev Cell ; 34(5): 493-504, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26321127

RESUMO

Postnatal organogenesis occurs in an immune competent environment and is tightly controlled by interplay between positive and negative regulators. Innate immune cells have beneficial roles in postnatal tissue remodeling, but roles for the adaptive immune system are currently unexplored. Here we show that adaptive immune responses participate in the normal postnatal development of a non-lymphoid epithelial tissue. Since the mammary gland (MG) is the only organ developing predominantly after birth, we utilized it as a powerful system to study adaptive immune regulation of organogenesis. We found that antigen-mediated interactions between mammary antigen-presenting cells and interferon-γ (IFNγ)-producing CD4+ T helper 1 cells participate in MG postnatal organogenesis as negative regulators, locally orchestrating epithelial rearrangement. IFNγ then affects luminal lineage differentiation. This function of adaptive immune responses, regulating normal development, changes the paradigm for studying players of postnatal organogenesis and provides insights into immune surveillance and cancer transformation.


Assuntos
Imunidade Adaptativa/imunologia , Células Apresentadoras de Antígenos/imunologia , Mama/imunologia , Células Epiteliais/citologia , Epitélio/metabolismo , Organogênese/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Mama/crescimento & desenvolvimento , Mama/metabolismo , Células Epiteliais/imunologia , Epitélio/imunologia , Feminino , Humanos , Imunidade Inata/imunologia , Interferon gama/metabolismo , Camundongos
9.
J Immunol ; 195(1): 87-95, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26002979

RESUMO

Acute respiratory distress syndrome (ARDS) is a devastating disorder characterized by increased alveolar permeability with no effective treatment beyond supportive care. Current mechanisms underlying ARDS focus on alveolar endothelial and epithelial injury caused by products of innate immune cells and platelets. However, the role of adaptive immune cells in ARDS remains largely unknown. In this study, we report that expansion of Ag-specific αßTh17 cells contributes to ARDS by local secretion of IL-17A, which in turn directly increases alveolar epithelial permeability. Mice with a highly restrictive defect in Ag-specific αßTh17 cells were protected from experimental ARDS induced by a single dose of endotracheal LPS. Loss of IL-17 receptor C or Ab blockade of IL-17A was similarly protective, further suggesting that IL-17A released by these cells was responsible for this effect. LPS induced a rapid and specific clonal expansion of αßTh17 cells in the lung, as determined by deep sequencing of the hypervariable CD3RßVJ region of the TCR. Our findings could be relevant to ARDS in humans, because we found significant elevation of IL-17A in bronchoalveolar lavage fluid from patients with ARDS, and rIL-17A directly increased permeability across cultured human alveolar epithelial monolayers. These results reveal a previously unexpected role for adaptive immune responses that increase alveolar permeability in ARDS and suggest that αßTh17 cells and IL-17A could be novel therapeutic targets for this currently untreatable disease.


Assuntos
Interleucina-17/imunologia , Alvéolos Pulmonares/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Síndrome do Desconforto Respiratório/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Animais , Anticorpos/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Humanos , Interleucina-17/antagonistas & inibidores , Interleucina-17/genética , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Transgênicos , Permeabilidade , Cultura Primária de Células , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Ratos , Ratos Sprague-Dawley , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/patologia , Células Th17/efeitos dos fármacos , Células Th17/patologia
10.
PLoS One ; 8(3): e58966, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23505568

RESUMO

Improper regulation of B cell responses leads to excessive production of antibodies and contributes to the development of autoimmune disease. T helper 17 (Th17) cells also drive the development of autoimmune disease, but the role of B cells in shaping Th17 cell-mediated immune responses, as well as the reciprocal regulation of B cell responses by IL-17 family cytokines, remains unclear. The aim of this study was to characterize the regulation of IL-17A and IL-17F in a model of T cell-dependent B cell activation. Stimulation of primary human B cell and peripheral blood mononuclear cell (BT) co-cultures with α-IgM and a non-mitogenic concentration of superantigens for three days promoted a Th17 cell response as evidenced by increased expression of Th17-related gene transcripts, including Il17f, Il21, Il22, and Il23r, in CD4 T cells, as well as the secretion of IL-17A and IL-17F protein. We tested the ability of 144 pharmacologic modulators representing 91 different targets or pathways to regulate IL-17A and IL-17F production in these stimulated BT co-cultures. IL-17A production was found to be preferentially sensitive to inhibition of the PI3K/mTOR pathway, while prostaglandin EP receptor agonists, including PGE2, increased IL-17A concentrations. In contrast, the production of IL-17F was inhibited by PGE2, but selectively increased by TLR2 and TLR5 agonists. These results indicate that IL-17A regulation is distinct from IL-17F in stimulated BT co-cultures and that this co-culture approach can be used to identify pathway mechanisms and novel agents that selectively inhibit production of IL-17A or IL-17F.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Interleucina-17/metabolismo , Ativação Linfocitária/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Calcitriol/farmacologia , Comunicação Celular , Técnicas de Cocultura , Humanos , Imunofenotipagem , Interleucina-17/genética , Fenótipo , Piperazinas/farmacologia , Cultura Primária de Células , Propanóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Células Th17/imunologia , Células Th17/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 304(7): G673-9, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23348803

RESUMO

Contraction of intestinal myofibroblasts (IMF) contributes to the development of strictures and fistulas seen in inflammatory bowel disease, but the mechanisms that regulate tension within these cells are poorly understood. In this study we investigated the role of nitric oxide (NO) signaling in C-type natriuretic peptide (CNP)-induced relaxation of IMF. We found that treatment with ODQ, a soluble guanylyl cyclase (sGC) inhibitor, or N(G)-nitro-L-arginine (L-NNA) or N(G)-monomethyl-L-arginine (L-NMMA), inhibitors of NO production, all impaired the relaxation of human and mouse IMF in response to CNP. ODQ, L-NNA, and L-NMMA also prevented CNP-induced elevations in cGMP concentrations, and L-NNA or L-NMMA blocked CNP-induced decreases in myosin light phosphorylation. IMF isolated from transgenic mice deficient in inducible nitric oxide synthase (iNOS) had reduced relaxation responses to CNP compared with IMF from control mice and were insensitive to the effects of ODQ, L-NNA, and L-NMMA on CNP treatment. Together these data indicate that stimulation of sGC though NO produced by iNOS activation is required for maximal CNP-induced relaxation in IMF.


Assuntos
Guanilato Ciclase/metabolismo , Miofibroblastos/fisiologia , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , GMP Cíclico/metabolismo , Guanilato Ciclase/antagonistas & inibidores , Humanos , Camundongos , Relaxamento Muscular/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Peptídeo Natriurético Tipo C/farmacologia , Nitroarginina/farmacologia , Oxidiazóis/farmacologia , Quinoxalinas/farmacologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Guanilil Ciclase Solúvel , ômega-N-Metilarginina/farmacologia
12.
Nat Med ; 18(4): 547-54, 2012 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-22388091

RESUMO

Emerging evidence suggests that the T helper 17 (T(H)17) subset of αß T cells contributes to the development of allergic asthma. In this study, we found that mice lacking the αvß8 integrin on dendritic cells did not generate T(H)17 cells in the lung and were protected from airway hyper-responsiveness in response to house dust mite and ovalbumin sensitization and challenge. Because loss of T(H)17 cells inhibited airway narrowing without any obvious effects on airway inflammation or epithelial morphology, we examined the direct effects of T(H)17 cytokines on mouse and human airway smooth muscle function. Interleukin-17A (IL-17A), but not IL-17F or IL-22, enhanced contractile force generation of airway smooth muscle through an IL-17 receptor A (IL-17RA)-IL-17RC, nuclear factor κ light-chain enhancer of activated B cells (NF-κB)-ras homolog gene family, member A (RhoA)-Rho-associated coiled-coil containing protein kinase 2 (ROCK2) signaling cascade. Mice lacking integrin αvß8 on dendritic cells showed impaired activation of this pathway after ovalbumin sensitization and challenge, and the diminished contraction of the tracheal rings in these mice was reversed by IL-17A. These data indicate that the IL-17A produced by T(H)17 cells contributes to allergen-induced airway hyper-responsiveness through direct effects on airway smooth muscle.


Assuntos
Asma/patologia , Interleucina-17/metabolismo , Interleucina-17/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Células Th17/metabolismo , Análise de Variância , Animais , Asma/imunologia , Antígeno CD11c/genética , Antígenos CD4 , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Técnicas In Vitro , Integrina alfaV/genética , Masculino , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agonistas Muscarínicos/farmacologia , Contração Muscular/fisiologia , Ovalbumina/imunologia , Cloreto de Potássio/farmacologia , Sistema Respiratório/citologia , Transdução de Sinais/efeitos dos fármacos , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Gastroenterology ; 141(5): 1802-12, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21723222

RESUMO

BACKGROUND & AIMS: The intestinal immune system is tightly regulated to prevent responses against the many nonpathogenic antigens in the gut. Transforming growth factor (TGF)-ß is a cytokine that maintains intestinal homeostasis, in part by inducing Foxp3(+) regulatory T cells (Tregs) that suppress immune responses. TGF-ß is expressed at high levels in the gastrointestinal tract as a latent complex that must be activated. However, the pathways that control TGF-ß activation in the intestine are poorly defined. We investigated the cellular and molecular pathways that control activation of TGF-ß and induction of Foxp3(+) Tregs in the intestines of mice to maintain immune homeostasis. METHODS: Subsets of intestinal dendritic cells (DCs) were examined for their capacity to activate TGF-ß and induce Foxp3(+) Tregs in vitro. Mice were fed oral antigen, and induction of Foxp3(+) Tregs was measured. RESULTS: A tolerogenic subset of intestinal DCs that express CD103 were specialized to activate latent TGF-ß, and induced Foxp3(+) Tregs independently of the vitamin A metabolite retinoic acid. The integrin αvß8, which activates TGF-ß, was significantly up-regulated on CD103(+) intestinal DCs. DCs that lack expression of integrin αvß8 had reduced ability to activate latent TGF-ß and induce Foxp3(+) Tregs in vitro and in vivo. CONCLUSIONS: CD103(+) intestinal DCs promote a tolerogenic environment in the intestines of mice via integrin αvß8-mediated activation of TGF-ß.


Assuntos
Células Dendríticas/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Integrinas/metabolismo , Intestinos/citologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos CD/metabolismo , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Homeostase/fisiologia , Sistema Imunitário/fisiologia , Técnicas In Vitro , Cadeias alfa de Integrinas/metabolismo , Integrinas/genética , Camundongos , Camundongos Knockout , Modelos Animais , Ovalbumina/farmacologia , Linfócitos T Reguladores/citologia , Tretinoína/metabolismo
14.
J Clin Invest ; 120(12): 4436-44, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21099117

RESUMO

Th17 cells promote a variety of autoimmune diseases, including psoriasis, multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. TGF-ß is required for conversion of naive T cells to Th17 cells, but the mechanisms regulating this process are unknown. Integrin αvß8 on DCs can activate TGF-ß, and this process contributes to the development of induced Tregs. Here, we have now shown that integrin αvß8 expression on DCs plays a critical role in the differentiation of Th17 cells. Th17 cells were nearly absent in the colons of mice lacking αvß8 expression on DCs. In addition, these mice and the DCs harvested from them had an impaired ability to convert naive T cells into Th17 cells in vivo and in vitro, respectively. Importantly, mice lacking αvß8 on DCs showed near-complete protection from experimental autoimmune encephalomyelitis. Our results therefore suggest that the integrin αvß8 pathway is biologically important and that αvß8 expression on DCs could be a therapeutic target for the treatment of Th17-driven autoimmune disease.


Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Integrinas/metabolismo , Células Th17/imunologia , Animais , Sequência de Bases , Diferenciação Celular/imunologia , Colo/imunologia , Colo/patologia , Citocinas/biossíntese , Citocinas/genética , Primers do DNA/genética , Células Dendríticas/patologia , Encefalomielite Autoimune Experimental/patologia , Expressão Gênica , Técnicas In Vitro , Integrinas/deficiência , Integrinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Th17/patologia
15.
J Clin Invest ; 119(12): 3713-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19884654

RESUMO

Milk fat globule epidermal growth factor 8 (Mfge8) is a soluble glycoprotein known to regulate inflammation and immunity by mediating apoptotic cell clearance. Since fibrosis can occur as a result of exaggerated apoptosis and inflammation, we set out to investigate the hypothesis that Mfge8 might negatively regulate tissue fibrosis. We report here that Mfge8 does decrease the severity of tissue fibrosis in a mouse model of pulmonary fibrosis; however, it does so not through effects on inflammation and apoptotic cell clearance, but by binding and targeting collagen for cellular uptake through its discoidin domains. Initial analysis revealed that Mfge8-/- mice exhibited enhanced pulmonary fibrosis after bleomycin-induced lung injury. However, they did not have increased inflammation or impaired apoptotic cell clearance after lung injury compared with Mfge8+/+ mice; rather, they had a defect in collagen turnover. Further experiments indicated that Mfge8 directly bound collagen and that Mfge8-/- macrophages exhibited defective collagen uptake that could be rescued by recombinant Mfge8 containing at least one discoidin domain. These data demonstrate a critical role for Mfge8 in decreasing the severity of murine tissue fibrosis by facilitating the removal of accumulated collagen.


Assuntos
Antígenos de Superfície/metabolismo , Colágeno/metabolismo , Macrófagos Alveolares/metabolismo , Proteínas do Leite/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Antígenos de Superfície/química , Antígenos de Superfície/genética , Apoptose , Sequência de Bases , Bleomicina/toxicidade , Primers do DNA/genética , Discoidinas , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Feminino , Lectinas/química , Lectinas/genética , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Leite/química , Proteínas do Leite/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
Am J Physiol Gastrointest Liver Physiol ; 293(6): G1272-80, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17947456

RESUMO

Chemotaxis (i.e., directed migration) of hepatic stellate cells to areas of inflammation is a requisite event in the liver's response to injury. Previous studies of signaling pathways that regulate stellate cell migration suggest a key role for focal adhesions, but the exact function of these protein complexes in motility remains unclear. Focal adhesions attach a cell to its substrate and therefore must be regulated in a highly coordinated manner during migration. To test the hypothesis that focal adhesion turnover is an essential early event for chemotaxis in stellate cells, we employed a live-cell imaging technique in which chemotaxis was induced by locally stimulating the tips of rat stellate cell protrusions with platelet-derived growth factor-BB (PDGF). Focal adhesions were visualized with an antibody directed against vinculin, a structural component of the focal adhesion complex. PDGF triggered rapid disassembly of adhesions within 6.25 min, subsequent reassembly by 12.5 min, and continued adhesion assembly in concert with the spreading protrusion until the completion of chemotaxis. Blockade of adhesion disassembly by growing cells on fibronectin or treatment with nocodazole prevented a chemotactic response to PDGF. Augmentation of adhesion disassembly with ML-7 enhanced the chemotactic response to PDGF. These data suggest that focal adhesion disassembly is an essential early event in stellate cell chemotaxis in response to PDGF.


Assuntos
Quimiotaxia/fisiologia , Adesões Focais/fisiologia , Hepatócitos/fisiologia , Fator de Crescimento Derivado de Plaquetas/administração & dosagem , Animais , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
17.
Nature ; 449(7160): 361-5, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17694047

RESUMO

The cytokine transforming growth factor-beta (TGF-beta) is an important negative regulator of adaptive immunity. TGF-beta is secreted by cells as an inactive precursor that must be activated to exert biological effects, but the mechanisms that regulate TGF-beta activation and function in the immune system are poorly understood. Here we show that conditional loss of the TGF-beta-activating integrin alpha(v)beta8 on leukocytes causes severe inflammatory bowel disease and age-related autoimmunity in mice. This autoimmune phenotype is largely due to lack of alpha(v)beta8 on dendritic cells, as mice lacking alpha(v)beta8 principally on dendritic cells develop identical immunological abnormalities as mice lacking alpha(v)beta8 on all leukocytes, whereas mice lacking alpha(v)beta8 on T cells alone are phenotypically normal. We further show that dendritic cells lacking alpha(v)beta8 fail to induce regulatory T cells (T(R) cells) in vitro, an effect that depends on TGF-beta activity. Furthermore, mice lacking alpha(v)beta8 on dendritic cells have reduced proportions of T(R) cells in colonic tissue. These results suggest that alpha(v)beta8-mediated TGF-beta activation by dendritic cells is essential for preventing immune dysfunction that results in inflammatory bowel disease and autoimmunity, effects that are due, at least in part, to the ability of alpha(v)beta8 on dendritic cells to induce and/or maintain tissue T(R) cells.


Assuntos
Autoimunidade/imunologia , Colite/metabolismo , Células Dendríticas/metabolismo , Integrinas/deficiência , Integrinas/metabolismo , Leucócitos/metabolismo , Envelhecimento/imunologia , Animais , Colite/imunologia , Colo/citologia , Colo/imunologia , Imunoglobulinas/sangue , Memória Imunológica , Integrinas/genética , Interferon gama/metabolismo , Interleucina-4/metabolismo , Leucócitos/imunologia , Leucócitos/patologia , Ativação Linfocitária , Contagem de Linfócitos , Camundongos , Fenótipo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Fator de Crescimento Transformador beta/metabolismo
18.
Hepatology ; 45(6): 1446-53, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17465006

RESUMO

UNLABELLED: Hepatic stellate cells play an essential role in the liver's injury response. Although stellate cells are defined by the presence of cytoplasmic protrusions, the function of these characteristic structures has been obscure. We hypothesized that stellate cell protrusions act by coupling injury-associated stimuli to chemotaxis. To test this hypothesis, we developed an assay for directly visualizing the response of living stellate cells in early primary culture to local stimulation of the tips of protrusions with platelet-derived growth factor-BB (PDGF). Stellate cells exhibited elongate protrusions containing actin, myosin, and tubulin. PDGF, but not cytochrome C, localized at a protrusion tip induced a coordinated series of morphological events--cell spreading at the tip, movement of the cell body toward the PDGF, and retraction of trailing protrusions--that resulted in chemotaxis. Soluble PDGF and AG 1296, a receptor tyrosine kinase inhibitor, both reduced stellate cell chemotaxis. PDGF-induced chemotaxis was associated with an early and transient increase in myosin phosphorylation within the spreading lamella. We observed that blebbistatin, a myosin II inhibitor, completely and reversibly blocked protrusion-mediated lamella formation and chemotaxis. Moreover, blockade of MRLC phosphorylation with the myosin light chain kinase inhibitor, ML-7, or the rho kinase inhibitor, Y-27632, blocked lamella formation, myosin phosphorylation within the protrusion, and chemotaxis. CONCLUSION: These results support a model in which protrusions permit stellate cells to promptly detect PDGF distant from their cell bodies and transduce this signal into mechanical forces that propel the cell toward the site of injury.


Assuntos
Indutores da Angiogênese/farmacologia , Quimiotaxia/fisiologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/ultraestrutura , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pseudópodes/fisiologia , Transdução de Sinais/fisiologia , Animais , Becaplermina , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Hepatócitos/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-sis , Pseudópodes/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
19.
Artigo em Inglês | MEDLINE | ID: mdl-16123199

RESUMO

The contractile force generated by hepatic stellate cells in response to endothelin-1 contributes to sinusoidal blood flow regulation and hepatic fibrosis. This study's aim was to directly test the widely held view that changes in cytosolic Ca2+ concentration ([Ca2+]i) mediate stellate cell force generation. Contractile force generation by primary cultures of rat hepatic stellate cells grown in three-dimensional collagen gels was directly and quantitatively measured using a force transducer. Stellate cell [Ca2+]i, myosin activation, and migration were quantified using standard techniques. [Ca2+]i was modulated using ionomycin, BAPTA, KCl, and removal of extracellular Ca2+. Removal of extracellular Ca2+ did not alter endothelin-1-stimulated force development or [Ca2+]i. Ionomycin, a Ca2+ ionophore, triggered an increase in [Ca2+]i that was three times greater than that stimulated by endothelin-1, but only induced 16% of the force and 38% of the myosin regulatory light chain (MLC) phosphorylation induced by endothelin-1. Physiological increases in [Ca2+]i induced by hyperkalemia had no effect on contractile force. Loading BAPTA, a Ca2+ chelator, in stellate cells completely blocked endothelin-1-induced increases in [Ca2+]i but had no effect on endothelin-1-stimulated force generation or MLC phosphorylation. In contrast, Y-27632, a selective rho-associated kinase inhibitor, inhibited endothelin-1-stimulated force generation by at least 70% and MLC phosphorylation by at least 80%. Taken together, these observations indicate that changes in [Ca2+]i are neither necessary nor sufficient for contractile force generation by rat stellate cells. Our results challenge the current model of contractile regulation in hepatic stellate cells and have important implications for our understanding of hepatic pathophysiology.


Assuntos
Cálcio/metabolismo , Endotelina-1/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Fígado/citologia , Fígado/efeitos dos fármacos , Animais , Células Cultivadas , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Cadeias Leves de Miosina/metabolismo , Miosinas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Transdução de Sinais , Quinases Associadas a rho
20.
Artigo em Inglês | MEDLINE | ID: mdl-12958024

RESUMO

Intestinal response to injury requires coordinated regulation of the tension exerted by subepithelial myofibroblasts (SEM). However, the signals governing relaxation of intestinal SEM have not been investigated. Our aim was to test the hypothesis that signal transduction pathways initiated by C-type natriuretic peptide (CNP) induce intestinal SEM relaxation. We directly quantified the effects of CNP on isometric tension exerted by cultured human colonic SEM. We also measured the effects of CNP on cGMP content, myosin regulatory light chain (MLC) phosphorylation, and cytosolic Ca2+ concentration. CNP induced relaxation of SEM within 10 s. By 10 min, relaxation reached a plateau that was sustained for 2 h. CNP-induced relaxation was saturable, with a maximal decrease in tension (51.7 +/- 3.8 dyn) observed at 250 nM. SEM relaxation in response to CNP constituted approximately 23% of total basal tension. CNP increased intracellular cGMP content and reduced MLC phosphorylation. Effects of CNP on cGMP and MLC exhibited the same dose dependence as CNP-induced relaxation. MLC phosphorylation decreased within 2 min of CNP exposure and was sustained for at least 45 min. CNP also stimulated a large transient increase in cytosolic Ca2+ concentration that occurred within 30 s and was nearly complete by 1 min. We also observed that calyculin-A, a potent inhibitor of MLC phosphatase, completely abolished the reduction in MLC phosphorylation induced by CNP. These results suggest that CNP induces intestinal SEM relaxation through cGMP-associated reductions in MLC phosphorylation. Moreover, these findings raise the possibility that CNP plays a role in intestinal wound healing.


Assuntos
Colo/efeitos dos fármacos , Peptídeo Natriurético Tipo C/farmacologia , Cálcio/metabolismo , Cálcio/fisiologia , Colo/citologia , GMP Cíclico/metabolismo , Citosol/enzimologia , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Humanos , Toxinas Marinhas , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Oxazóis/farmacologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA