Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 620(7975): 881-889, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37558878

RESUMO

Dendritic cells (DCs) have a role in the development and activation of self-reactive pathogenic T cells1,2. Genetic variants that are associated with the function of DCs have been linked to autoimmune disorders3,4, and DCs are therefore attractive therapeutic targets for such diseases. However, developing DC-targeted therapies for autoimmunity requires identification of the mechanisms that regulate DC function. Here, using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies, we identify a regulatory loop of negative feedback that operates in DCs to limit immunopathology. Specifically, we find that lactate, produced by activated DCs and other immune cells, boosts the expression of NDUFA4L2 through a mechanism mediated by hypoxia-inducible factor 1α (HIF-1α). NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs that are involved in the control of pathogenic autoimmune T cells. We also engineer a probiotic that produces lactate and suppresses T cell autoimmunity through the activation of HIF-1α-NDUFA4L2 signalling in DCs. In summary, we identify an immunometabolic pathway that regulates DC function, and develop a synthetic probiotic for its therapeutic activation.


Assuntos
Doenças Autoimunes , Sistema Nervoso Central , Células Dendríticas , Subunidade alfa do Fator 1 Induzível por Hipóxia , Ácido Láctico , Humanos , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/prevenção & controle , Autoimunidade , Sistema Nervoso Central/citologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ácido Láctico/metabolismo , Probióticos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/imunologia , Retroalimentação Fisiológica , Lactase/genética , Lactase/metabolismo , Análise de Célula Única
2.
Nature ; 598(7882): 662-666, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34616044

RESUMO

The availability of L-arginine in tumours is a key determinant of an efficient anti-tumour T cell response1-4. Consequently, increases of typically low L-arginine concentrations within the tumour may greatly potentiate the anti-tumour responses of immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1)-blocking antibodies5. However, currently no means are available to locally increase intratumoural L-arginine levels. Here we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumours and continuously converts ammonia, a metabolic waste product that accumulates in tumours6, to L-arginine. Colonization of tumours with these bacteria increased intratumoural L-arginine concentrations, increased the number of tumour-infiltrating T cells and had marked synergistic effects with PD-L1 blocking antibodies in the clearance of tumours. The anti-tumour effect of these bacteria was mediated by L-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumour microenvironment leading to enhanced efficacy of immunotherapies.


Assuntos
Imunoterapia/métodos , Engenharia Metabólica , Microrganismos Geneticamente Modificados , Neoplasias Experimentais/terapia , Transferência Adotiva , Animais , Arginina/metabolismo , Antígeno B7-H1/antagonistas & inibidores , Linhagem Celular Tumoral , Escherichia coli , Feminino , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/microbiologia , Probióticos , Proteoma , Biologia Sintética , Linfócitos T/imunologia , Microambiente Tumoral/imunologia
3.
J Biol Chem ; 296: 100733, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33957124

RESUMO

A disintegrin and metalloprotease 17 (ADAM17) is a cell-surface metalloprotease that serves as the principle sheddase for tumor necrosis factor α (TNFα), interleukin-6 receptor (IL-6R), and several ligands of the epidermal growth factor receptor (EGFR), regulating these crucial signaling pathways. ADAM17 activation requires its transmembrane domain, but not its cytoplasmic domain, and little is known about the role of this domain in vivo. To investigate, we used CRISPR-Cas9 to mutate the endogenous Adam17 locus in mice to produce a mutant ADAM17 lacking its cytoplasmic domain (Adam17Δcyto). Homozygous Adam17Δcyto animals were born at a Mendelian ratio and survived into adulthood with slightly wavy hair and curled whiskers, consistent with defects in ADAM17/EGFR signaling. At birth, Adam17Δcyto mice resembled Adam17-/- mice in that they had open eyes and enlarged semilunar heart valves, but they did not have bone growth plate defects. The deletion of the cytoplasmic domain resulted in strongly decreased ADAM17 protein levels in all tissues and cells examined, providing a likely cause for the hypomorphic phenotype. In functional assays, Adam17Δcyto mouse embryonic fibroblasts and bone-marrow-derived macrophages had strongly reduced ADAM17 activity, consistent with the reduced protein levels. Nevertheless, ADAM17Δcyto could be stimulated by PMA, a well-characterized posttranslational activator of ADAM17, corroborating that the cytoplasmic domain of endogenous ADAM17 is not required for its rapid response to PMA. Taken together, these results provide the first evidence that the cytoplasmic domain of ADAM17 plays a pivotal role in vivo in regulating ADAM17 levels and function.


Assuntos
Proteína ADAM17/química , Proteína ADAM17/metabolismo , Citoplasma/metabolismo , Proteína ADAM17/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Feminino , Fibroblastos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Domínios Proteicos , Estabilidade Proteica , Deleção de Sequência
4.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673337

RESUMO

Notch signaling is critical for controlling a variety of cell fate decisions during metazoan development and homeostasis. This unique, highly conserved signaling pathway relies on cell-to-cell contact, which triggers the proteolytic release of the cytoplasmic domain of the membrane-anchored transcription factor Notch from the membrane. A disintegrin and metalloproteinase (ADAM) proteins are crucial for Notch activation by processing its S2 site. While ADAM10 cleaves Notch1 under physiological, ligand-dependent conditions, ADAM17 mainly cleaves Notch1 under ligand-independent conditions. However, the mechanism(s) that regulate the distinct contributions of these ADAMs in Notch processing remain unclear. Using cell-based assays in mouse embryonic fibroblasts (mEFs) lacking ADAM10 and/or ADAM17, we aimed to clarify what determines the relative contributions of ADAM10 and ADAM17 to ligand-dependent or ligand-independent Notch processing. We found that EDTA-stimulated ADAM17-dependent Notch1 processing is rapid and requires the ADAM17-regulators iRhom1 and iRhom2, whereas the Delta-like 4-induced ligand-dependent Notch1 processing is slower and requires ADAM10. The selectivity of ADAM17 for EDTA-induced Notch1 processing can most likely be explained by a preference for ADAM17 over ADAM10 for the Notch1 cleavage site and by the stronger inhibition of ADAM10 by EDTA. The physiological ADAM10-dependent processing of Notch1 cannot be compensated for by ADAM17 in Adam10-/- mEFs, or by other ADAMs shown here to be able to cleave the Notch1 cleavage site, such as ADAMs9, 12, and 19. Collectively, these results provide new insights into the mechanisms underlying the substrate selectivity of ADAM10 and ADAM17 towards Notch1.


Assuntos
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas de Membrana/metabolismo , Proteólise , Receptor Notch1/metabolismo , Proteína ADAM10/genética , Proteína ADAM17/genética , Secretases da Proteína Precursora do Amiloide/genética , Animais , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Receptor Notch1/genética
5.
J Biol Chem ; 291(25): 13014-27, 2016 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-27056325

RESUMO

Covalent modification of histones is a fundamental mechanism of regulated gene expression in eukaryotes, and interpretation of histone modifications is an essential feature of epigenetic control. Bromodomains are specialized binding modules that interact with acetylated histones, linking chromatin recognition to gene transcription. Because of their ability to function in a domain-specific fashion, selective disruption of bromodomain:acetylated histone interactions with chemical probes serves as a powerful means for understanding biological processes regulated by these chromatin adaptors. Here we describe the discovery and characterization of potent and selective small molecule inhibitors for the bromodomains of CREBBP/EP300 that engage their target in cellular assays. We use these tools to demonstrate a critical role for CREBBP/EP300 bromodomains in regulatory T cell biology. Because regulatory T cell recruitment to tumors is a major mechanism of immune evasion by cancer cells, our data highlight the importance of CREBBP/EP300 bromodomain inhibition as a novel, small molecule-based approach for cancer immunotherapy.


Assuntos
Proteína de Ligação a CREB/antagonistas & inibidores , Proteína p300 Associada a E1A/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Proteína de Ligação a CREB/química , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Proteína p300 Associada a E1A/química , Proteína p300 Associada a E1A/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Histonas/metabolismo , Humanos , Simulação de Acoplamento Molecular , Estrutura Terciária de Proteína/efeitos dos fármacos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Transcriptoma/efeitos dos fármacos
6.
Nature ; 468(7327): 1119-23, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21068722

RESUMO

Interaction of pathogens with cells of the immune system results in activation of inflammatory gene expression. This response, although vital for immune defence, is frequently deleterious to the host due to the exaggerated production of inflammatory proteins. The scope of inflammatory responses reflects the activation state of signalling proteins upstream of inflammatory genes as well as signal-induced assembly of nuclear chromatin complexes that support mRNA expression. Recognition of post-translationally modified histones by nuclear proteins that initiate mRNA transcription and support mRNA elongation is a critical step in the regulation of gene expression. Here we present a novel pharmacological approach that targets inflammatory gene expression by interfering with the recognition of acetylated histones by the bromodomain and extra terminal domain (BET) family of proteins. We describe a synthetic compound (I-BET) that by 'mimicking' acetylated histones disrupts chromatin complexes responsible for the expression of key inflammatory genes in activated macrophages, and confers protection against lipopolysaccharide-induced endotoxic shock and bacteria-induced sepsis. Our findings suggest that synthetic compounds specifically targeting proteins that recognize post-translationally modified histones can serve as a new generation of immunomodulatory drugs.


Assuntos
Anti-Inflamatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Inflamação , Macrófagos/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Benzodiazepinas , Células Cultivadas , Epigenômica , Estudo de Associação Genômica Ampla , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Estimativa de Kaplan-Meier , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Infecções por Salmonella/tratamento farmacológico , Infecções por Salmonella/imunologia , Infecções por Salmonella/fisiopatologia , Infecções por Salmonella/prevenção & controle , Salmonella typhimurium , Sepse/tratamento farmacológico , Sepse/prevenção & controle , Choque Séptico/tratamento farmacológico , Choque Séptico/prevenção & controle
7.
Drug Discov Today Technol ; 19: 39-44, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27769356

RESUMO

Epigenetic control of gene expression is enforced in part through histone modifications. Bromodomain and extra terminal domain (BET) proteins function as crucial chromatin readers, responsible for interpretation of the chromatin code in diverse cellular contexts, ultimately impacting gene transcription. BET proteins can play a major role in inflammation by profoundly affecting the biology of the Thelper 17 (TH17) lineage. We summarize recent studies focusing on BET inhibition as a viable therapeutic alternative for the control of autoimmune diseases driven by aberrant activation of TH17 cells.


Assuntos
Proteínas Nucleares/antagonistas & inibidores , Domínios Proteicos , Células Th17/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Animais , Diferenciação Celular , Citocinas/metabolismo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Células Th17/citologia , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
8.
Elife ; 132024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38860651

RESUMO

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.


Assuntos
Proteína ADAM17 , Células de Langerhans , Lúpus Eritematoso Sistêmico , Pele , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Animais , Humanos , Células de Langerhans/metabolismo , Camundongos , Pele/metabolismo , Pele/patologia , Pele/efeitos da radiação , Lúpus Eritematoso Sistêmico/metabolismo , Raios Ultravioleta/efeitos adversos , Feminino , Modelos Animais de Doenças , Transtornos de Fotossensibilidade/metabolismo , Interferons/metabolismo , Camundongos Endogâmicos MRL lpr
9.
bioRxiv ; 2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36993446

RESUMO

Dendritic cells (DCs) control the generation of self-reactive pathogenic T cells. Thus, DCs are considered attractive therapeutic targets for autoimmune diseases. Using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies we identified a negative feedback regulatory pathway that operates in DCs to limit immunopathology. Specifically, we found that lactate, produced by activated DCs and other immune cells, boosts NDUFA4L2 expression through a mechanism mediated by HIF-1α. NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs involved in the control of pathogenic autoimmune T cells. Moreover, we engineered a probiotic that produces lactate and suppresses T-cell autoimmunity in the central nervous system via the activation of HIF-1α/NDUFA4L2 signaling in DCs. In summary, we identified an immunometabolic pathway that regulates DC function, and developed a synthetic probiotic for its therapeutic activation.

10.
Nat Rev Drug Discov ; 20(12): 941-960, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34616030

RESUMO

The steadfast advance of the synthetic biology field has enabled scientists to use genetically engineered cells, instead of small molecules or biologics, as the basis for the development of novel therapeutics. Cells endowed with synthetic gene circuits can control the localization, timing and dosage of therapeutic activities in response to specific disease biomarkers and thus represent a powerful new weapon in the fight against disease. Here, we conceptualize how synthetic biology approaches can be applied to programme living cells with therapeutic functions and discuss the advantages that they offer over conventional therapies in terms of flexibility, specificity and predictability, as well as challenges for their development. We present notable advances in the creation of engineered cells that harbour synthetic gene circuits capable of biological sensing and computation of signals derived from intracellular or extracellular biomarkers. We categorize and describe these developments based on the cell scaffold (human or microbial) and the site at which the engineered cell exerts its therapeutic function within its human host. The design of cell-based therapeutics with synthetic biology is a rapidly growing strategy in medicine that holds great promise for the development of effective treatments for a wide variety of human diseases.


Assuntos
Engenharia Celular/métodos , Engenharia Genética/mortalidade , Biologia Sintética , Terapia Baseada em Transplante de Células e Tecidos/tendências , Redes Reguladoras de Genes , Engenharia Genética/métodos , Humanos , Biologia Sintética/métodos , Biologia Sintética/tendências
11.
Ann N Y Acad Sci ; 1506(1): 98-117, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34786712

RESUMO

Synthetic biology has the potential to transform cell- and gene-based therapies for a variety of diseases. Sophisticated tools are now available for both eukaryotic and prokaryotic cells to engineer cells to selectively achieve therapeutic effects in response to one or more disease-related signals, thus sparing healthy tissue from potentially cytotoxic effects. This report summarizes the Keystone eSymposium "Synthetic Biology: At the Crossroads of Genetic Engineering and Human Therapeutics," which took place on May 3 and 4, 2021. Given that several therapies engineered using synthetic biology have entered clinical trials, there was a clear need for a synthetic biology symposium that emphasizes the therapeutic applications of synthetic biology as opposed to the technical aspects. Presenters discussed the use of synthetic biology to improve T cell, gene, and viral therapies, to engineer probiotics, and to expand upon existing modalities and functions of cell-based therapies.


Assuntos
Congressos como Assunto/tendências , Engenharia Genética/tendências , Terapia Genética/tendências , Relatório de Pesquisa , Biologia Sintética/tendências , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Marcação de Genes/métodos , Marcação de Genes/tendências , Engenharia Genética/métodos , Terapia Genética/métodos , Humanos , Células Matadoras Naturais/imunologia , Aprendizado de Máquina/tendências , Biologia Sintética/métodos , Linfócitos T/imunologia
12.
Prostaglandins Other Lipid Mediat ; 92(1-4): 33-43, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20214997

RESUMO

Asthma, chronic obstructive pulmonary disease (COPD) and acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are characterized by neutrophilic inflammation and elevated levels of leukotriene B4 (LTB4). However, the exact role of LTB4 pathways in mediating pulmonary neutrophilia and the potential therapeutic application of LTB4 receptor antagonists in these diseases remains controversial. Here we show that a novel dual BLT1 and BLT2 receptor antagonist, RO5101576, potently inhibited LTB4-evoked calcium mobilization in HL-60 cells and chemotaxis of human neutrophils. RO5101576 significantly attenuated LTB4-evoked pulmonary eosinophilia in guinea pigs. In non-human primates, RO5101576 inhibited allergen and ozone-evoked pulmonary neutrophilia, with comparable efficacy to budesonide (allergic responses). RO5101576 had no effects on LPS-evoked neutrophilia in guinea pigs and cigarette smoke-evoked neutrophilia in mice and rats. In toxicology studies RO5101576 was well-tolerated. Theses studies show differential effects of LTB4 receptor antagonism on neutrophil responses in vivo and suggest RO5101576 may represent a potential new treatment for pulmonary neutrophilia in asthma.


Assuntos
Benzodioxóis/farmacologia , Fenilpropionatos/farmacologia , Pneumonia/tratamento farmacológico , Primatas , Receptores do Leucotrieno B4/antagonistas & inibidores , Animais , Benzodioxóis/uso terapêutico , Benzodioxóis/toxicidade , Cães , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Feminino , Cobaias , Células HL-60 , Humanos , Hipersensibilidade/complicações , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Ozônio/farmacologia , Fenilpropionatos/uso terapêutico , Fenilpropionatos/toxicidade , Pneumonia/induzido quimicamente , Pneumonia/complicações , Pneumonia/metabolismo , Ratos , Receptores do Leucotrieno B4/metabolismo , Fumar/efeitos adversos , Testes de Toxicidade
13.
Nat Commun ; 11(1): 2739, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483165

RESUMO

Synthetic biology is a powerful tool to create therapeutics which can be rationally designed to enable unique and combinatorial functionalities. Here we utilize non-pathogenic E coli Nissle as a versatile platform for the development of a living biotherapeutic for the treatment of cancer. The engineered bacterial strain, referred to as SYNB1891, targets STING-activation to phagocytic antigen-presenting cells (APCs) in the tumor and activates complementary innate immune pathways. SYNB1891 treatment results in efficacious antitumor immunity with the formation of immunological memory in murine tumor models and robust activation of human APCs. SYNB1891 is designed to meet manufacturability and regulatory requirements with built in biocontainment features which do not compromise its efficacy. This work provides a roadmap for the development of future therapeutics and demonstrates the transformative potential of synthetic biology for the treatment of human disease when drug development criteria are incorporated into the design process for a living medicine.


Assuntos
Escherichia coli/imunologia , Imunoterapia/métodos , Proteínas de Membrana/imunologia , Neoplasias/terapia , Transdução de Sinais/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia Genética/métodos , Humanos , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/genética , Neoplasias/imunologia , Fagócitos/imunologia , Fagócitos/metabolismo , Transdução de Sinais/genética , Biologia Sintética/métodos , Biologia Sintética/tendências
14.
J Clin Invest ; 115(12): 3460-72, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16284650

RESUMO

Th1 inflammation and remodeling characterized by tissue destruction frequently coexist in human diseases. To further understand the mechanisms of these responses, we defined the role(s) of CCR5 in the pathogenesis of IFN-gamma-induced inflammation and remodeling in a murine emphysema model. IFN-gamma was a potent stimulator of the CCR5 ligands macrophage inflammatory protein-1alpha/CCL-3 (MIP-1alpha/CCL-3), MIP-1beta/CCL-4, and RANTES/CCL-5, among others. Antibody neutralization or null mutation of CCR5 decreased IFN-gamma-induced inflammation, DNA injury, apoptosis, and alveolar remodeling. These interventions decreased the expression of select chemokines, including CCR5 ligands and MMP-9, and increased levels of secretory leukocyte protease inhibitor. They also decreased the expression and/or activation of Fas, FasL, TNF, caspase-3, -8, and -9, Bid, and Bax. In accordance with these findings, cigarette smoke induced pulmonary inflammation, DNA injury, apoptosis, and emphysema via an IFN-gamma-dependent pathway(s), and a null mutation of CCR5 decreased these responses. These studies demonstrate that IFN-gamma is a potent stimulator of CC and CXC chemokines and highlight the importance of CCR5 in the pathogenesis of IFN-gamma-induced and cigarette smoke-induced inflammation, tissue remodeling, and emphysema. They also demonstrate that CCR5 is required for optimal IFN-gamma stimulation of its own ligands, other chemokines, MMPs, caspases, and cell death regulators and the inhibition of antiproteases.


Assuntos
Enfisema/patologia , Interferon gama/metabolismo , Receptores CCR5/fisiologia , Fumar , Animais , Anexina A5/química , Apoptose , Lavagem Broncoalveolar , Morte Celular , Quimiocinas/metabolismo , DNA/metabolismo , Primers do DNA/química , Enfisema/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação , Ligantes , Pulmão/metabolismo , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fenótipo , Alvéolos Pulmonares/metabolismo , RNA Mensageiro/metabolismo , Receptores CCR5/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
15.
J Pharmacol Exp Ther ; 327(3): 926-33, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18753410

RESUMO

Chemokine CC motif receptor (CCR) 5 is a major drug target for both inflammation and virology indications. The primary function of CCR5 is to mediate the trafficking of CCR5-expressing lymphocytes to any of the CCR5 ligands, which are often increased during inflammatory responses. In addition, CCR5 is a coreceptor for HIV, mediating R5 tropic HIV infection of CCR5-expressing CD4 T cells. We report the use of a novel method to assay the pharmacodynamic (PD) properties of small-molecule and antibody inhibitors of CCR5 ligand-induced activation by measuring phosphorylation of serine residue 349 in the cytoplasmic tail of human CCR5 using phosphoflow cytometry in whole blood. This assay is highly specific and measures CCR5 phosphorylation in both CD8(+) and CD4(+) T cells and allows the calculation of inhibitor IC(50) values from both lymphocyte subsets in the presence of CCR5 antagonists. In addition, this assay is cross-reactive to nonhuman primates and allows PD analysis in whole blood from rhesus and cynomolgus macaque. Using this assay, we identified different ligand-dependent response properties between CD8(+) and CD4(+) T cells, although CCR5 antagonists behave with similar properties against both cell types. The use of this assay may be of particular benefit to monitor PD effects of CCR5 inhibitors during drug development, preclinical in vivo studies, and in patients currently being treated for HIV or CCR5-mediated inflammatory diseases with CCR5 inhibitors. Similar phosphoflow approaches to other GPCR targets on circulating lymphocytes may prove to be the most reliable PD assay for preclinical and potentially clinical development.


Assuntos
Antagonistas dos Receptores CCR5 , Linfócitos T CD4-Positivos/química , Linfócitos T CD8-Positivos/química , Monitoramento de Medicamentos/métodos , Receptores CCR5/sangue , Animais , Citometria de Fluxo/métodos , Humanos , Concentração Inibidora 50 , Ligantes , Macaca , Macaca mulatta , Métodos , Farmacocinética , Fosforilação
16.
Sci Rep ; 7: 43139, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28233801

RESUMO

Atopic asthma is a prevalent respiratory disease that is characterized by inflammation, mucus hypersecretion, and airway hyperresponsiveness. The complexity of this heterogeneous disorder has commanded the need to better define asthma phenotypes based on underlying molecular mechanisms of disease. Although classically viewed as a type 2-regulated disease, type 17 helper T (Th17) cells are known to be influential in asthma pathogenesis, predominantly in asthmatics with neutrophilia and severe refractory disease. Bromodomain and extra-terminal domain (BET) chromatin adaptors serve as immunomodulators by directly regulating Th17 responses and Th17-mediated pathology in murine models of autoimmunity and infection. Based on this, we hypothesized that BET proteins may also play an essential role in neutrophil-dominant allergic airway disease. Using a murine model of neutrophil-dominant allergic airway disease, we demonstrate that BET inhibition limits pulmonary inflammation and alters the Th17-related inflammatory milieu in the lungs. In addition, inhibition of BET proteins improved lung function (specifically quasi-static lung compliance and tissue elastance) and reduced mucus production in airways. Overall, these studies show that BET proteins may have a critical role in asthma pathogenesis by altering type 17 inflammation, and thus interfering with BET-dependent chromatin signaling may provide clinical benefits to patients suffering from asthma.


Assuntos
Asma/patologia , Neutrófilos/imunologia , Fatores de Transcrição/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Inflamação/patologia , Pulmão/patologia , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/metabolismo , Células Th17/imunologia , Fatores de Transcrição/metabolismo
17.
J Leukoc Biol ; 78(2): 426-34, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15908459

RESUMO

Macrophages exist as sentinels in innate immune response and react by expressing proinflammatory cytokines and up-regulating antigen-presenting and costimulatory molecules. We report a novel function for prokineticin-1 (PK1)/endocrine gland-derived vascular endothelial growth factor. Screening of murine tissue sections and cells for specific binding site leads to the identification of macrophages as an in vivo cellular target for PK1. We demonstrate PK1 induces differentiation of murine and human bone marrow cells into the monocyte/macrophage lineage. Human peripheral blood monocytes respond to PK1 by morphological changes and down-regulation of B7-1, CD14, CC chemokine receptor 5, and CXC chemokine receptor 4. Monocytes treated with PK1 have elevated interleukin (IL)-12 and tumor necrosis factor alpha and down-regulated IL-10 production in response to lipopolysaccharide. PK1 induces a distinct monocyte-derived cell population, which is primed for release of proinflammatory cytokines that favor a T helper cell type 1 response.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Macrófagos/fisiologia , Células Progenitoras Mieloides/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/farmacologia , Animais , Antígeno B7-1/biossíntese , Células CHO , Diferenciação Celular/fisiologia , Cricetinae , Cricetulus , Citocinas/biossíntese , Humanos , Receptores de Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Camundongos , Receptores CCR5/biossíntese , Receptores CXCR4/biossíntese , Células Th1/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
18.
Microbes Infect ; 18(11): 675-686, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27416748

RESUMO

Chagasic cardiomyopathy, which is seen in Chagas disease, is the most severe and life-threatening manifestation of infection by the kinetoplastid Trypanosoma cruzi. Adipose tissue and diet play a major role in maintaining lipid homeostasis and regulating cardiac pathogenesis during the development of Chagas cardiomyopathy. We have previously reported that T. cruzi has a high affinity for lipoproteins and that the invasion rate of this parasite increases in the presence of cholesterol, suggesting that drugs that inhibit cholesterol synthesis, such as statins, could affect infection and the development of Chagasic cardiomyopathy. The dual epidemic of diabetes and obesity in Latin America, the endemic regions for Chagas disease, has led to many patients in the endemic region of infection having hyperlipidemia that is being treated with statins such as atorvastatin. The current study was performed to examine mice fed on either regular or high fat diet for effects of atorvastatin on T. cruzi infection-induced myocarditis and to evaluate the effect of this treatment during infection on adipose tissue physiology and cardiac pathology. Atorvastatin was found to regulate lipolysis and cardiac lipidopathy during acute T. cruzi infection in mice and to enhance tissue parasite load, cardiac LDL levels, inflammation, and mortality in during acute infection. Overall, these data suggest that statins, such as atorvastatin, have deleterious effects during acute Chagas disease.


Assuntos
Anticolesterolemiantes/administração & dosagem , Atorvastatina/administração & dosagem , Cardiomiopatia Chagásica/mortalidade , Dieta Hiperlipídica/efeitos adversos , Animais , Anticolesterolemiantes/efeitos adversos , Atorvastatina/efeitos adversos , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C3H , Análise de Sobrevida , Resultado do Tratamento
19.
Acta otorrinolaringol. cir. cuello (En línea) ; 49(1): 43-51, 2021. ilus, tab, graf
Artigo em Espanhol | COLNAL, LILACS | ID: biblio-1151895

RESUMO

Introducción: la dilatación endoscópica con balón ha mostrado ser una herramienta terapéutica y efectiva para el tratamiento de la disfunción de la trompa de Eustaquio. Al momento se desconocen los resultados en la población colombiana. Objetivos: determinar la mejoría clínica de la dilatación de la trompa de Eustaquio en pacientes adultos con disfunción tubárica crónica, en un centro de IV nivel de atención en Bogotá. Diseño: estudio observacional retrospectivo con componente analítico. Métodos: se revisaron los registros clínicos de pacientes intervenidos con la dilatación endoscópica de la trompa de Eustaquio; se describen los cambios objetivos y subjetivos en el posoperatorio. Resultados: se obtuvieron 22 pacientes entre los 18 y 66 años con seguimiento de 6 a 22 meses. Al 84,5 % se les realizó la intervención bilateral y al 15,8 % la unilateral, para un total de 38 oídos intervenidos. El 60 % lograron la normalización del timpanograma. Hubo una mejoría de las presiones del oído medio de -102,5 a -3,5 daPa (p = 0,005). Se alcanzó una mejoría estadísticamente significativa en 6/7 síntomas según la escala de severidad y reducción del gap aéreo-óseo en 7 dB y 4 dB del promedio de tonos puros por vía aérea (PTA) (p = 0,249; p = 0,711). Además, se generó un impacto positivo promedio de +32 en la escala de calidad de vida según el Glasgow Benefit Inventory (GBI). Conclusión: la dilatación de trompa de Eustaquio resulta ser una terapéutica segura y eficaz en población adulta, con mejoras estadísticamente significativas en la severidad de los síntomas y en las presiones del oído medio, lo cual genera un impacto positivo en la escala de calidad de vida según el GBI.


Introduction: Endoscopic balloon dilation has been shown to be a therapeutic and effective tool for the treatment of eustachian tube dysfunction. Now, the results in the Colombian population are unknown. Aim: Describe the clinical improvement of eustachian tube dilation in adult patients with chronic tubal dysfunction, in a 4th level complexity hospital in Bogotá. Design: Retrospective observational study with analytical component. Methods: The clinical records of patients undergoing endoscopic dilatation of the Eustachian tube were reviewed. Objective and subjective changes in the postoperative period are described. Results: 22 patients were obtained between 18 and 66 years and follow-up from 6 to 22 months. 84.5 % underwent bilateral intervention and 15.8 % unilaterally for a total of 38 operated ears. 60 % achieved tympanogram normalization. There was improvement of middle ear pressures from -102.5 to -3.5 daPa (p = 0.005). Statistically significant improvement in 6/7 symptoms according to the severity scale and reduction of air-bone Gap in 7 dB and 4 dB of airway PTA (p = 0.249; p = 0.711). Generating an average positive impact of +32 on the quality-of-life scale according to the Glasgow Benefit Inventory (GBI). Conclusions: Eustachian tube dilation proves to be a safe and effective therapy in an adult population with statistically significant improvements in the severity of symptoms, and in middle ear pressures. Generating a positive impact on the scale of quality of life according to the GBI.


Assuntos
Humanos , Masculino , Feminino , Adolescente , Adulto , Pessoa de Meia-Idade , Idoso , Adulto Jovem , Otopatias/cirurgia , Tuba Auditiva/cirurgia , Qualidade de Vida , Doença Crônica , Estudos Retrospectivos , Seguimentos , Resultado do Tratamento , Dilatação , Otopatias/diagnóstico
20.
JCI Insight ; 1(11)2016 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-27517095

RESUMO

Significant morbidity in cystic fibrosis (CF) results from chronic lung inflammation, most commonly due to Pseudomonas aeruginosa infection. Recent data suggest that IL-17 contributes to pathological inflammation in the setting of abnormal mucosal immunity, and type 17 immunity-driven inflammatory responses may represent a target to block aberrant inflammation in CF. Indeed, transcriptomic analysis of the airway epithelium from CF patients undergoing clinical bronchoscopy revealed upregulation of IL-17 downstream signature genes, implicating a substantial contribution of IL-17-mediated immunity in CF lungs. Bromodomain and extraterminal domain (BET) chromatin modulators can regulate T cell responses, specifically Th17-mediated inflammation, by mechanisms that include bromodomain-dependent inhibition of acetylated histones at the IL17 locus. Here, we show that, in vitro, BET inhibition potently suppressed Th17 cell responses in explanted CF tissue and inhibited IL-17-driven chemokine production in human bronchial epithelial cells. In an acute P. aeruginosa lung infection murine model, BET inhibition decreased inflammation, without exacerbating infection, suggesting that BET inhibition may be a potential therapeutic target in patients with CF.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA