Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
FASEB J ; 37(2): e22753, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36624683

RESUMO

Fibroblasts are stromal cells abundant throughout tissues, including the lungs. Fibroblasts are integral coordinators of immune cell recruitment through chemokine secretion. Circadian rhythms direct the recruitment of immune cells to the lung, which in turn impacts response to infection and survival. Although fibroblasts display robust circadian rhythms, the contribution of the fibroblast molecular clock to lung-specific migration of immune cells and recruitment remains to be established. Mice challenged intranasally with lipopolysaccharide (LPS) at dusk showed increased expression of the pro-inflammatory cytokine IL-1ß and chemokine CXCL5 in the lung, which was accompanied by increased neutrophil recruitment. Primary lung fibroblasts with knockdown of the core clock gene Bmal1 and immortalized Bmal1-/- lung fibroblasts also displayed increased Cxcl5 expression under IL-1ß stimulation. Conditioned media obtained from IL-1ß-stimulated Bmal1-/- immortalized fibroblasts-induced greater neutrophil migration compared with Bmal1+/+ lung fibroblast controls. Phosphorylation of the NF-κB subunit, p65, was enhanced in IL-1ß-stimulated Bmal1-/- lung fibroblasts, and pharmacological inhibition of NF-κB attenuated the enhanced CXCL5 production and neutrophil recruitment observed in these cells. Collectively, these results demonstrate that Bmal1 represses NF-κB activity in lung fibroblasts to control chemokine expression and immune cell recruitment during an inflammatory response.


Assuntos
Fatores de Transcrição ARNTL , NF-kappa B , Animais , Camundongos , Infiltração de Neutrófilos , Fatores de Transcrição ARNTL/genética , Fibroblastos , Movimento Celular , Ritmo Circadiano
2.
Nat Commun ; 13(1): 7217, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36470865

RESUMO

Dendritic cells play a key role in processing and presenting antigens to naïve T cells to prime adaptive immunity. Circadian rhythms are known to regulate many aspects of immunity; however, the role of circadian rhythms in dendritic cell function is still unclear. Here, we show greater T cell responses when mice are immunised in the middle of their rest versus their active phase. We find a circadian rhythm in antigen processing that correlates with rhythms in both mitochondrial morphology and metabolism, dependent on the molecular clock gene, Bmal1. Using Mdivi-1, a compound that promotes mitochondrial fusion, we are able to rescue the circadian deficit in antigen processing and mechanistically link mitochondrial morphology and antigen processing. Furthermore, we find that circadian changes in mitochondrial Ca2+ are central to the circadian regulation of antigen processing. Our results indicate that rhythmic changes in mitochondrial calcium, which are associated with changes in mitochondrial morphology, regulate antigen processing.


Assuntos
Relógios Circadianos , Camundongos , Animais , Relógios Circadianos/genética , Apresentação de Antígeno , Linfócitos T , Ritmo Circadiano/fisiologia , Antígenos , Vacinação , Células Dendríticas , Proteínas CLOCK/genética , Fatores de Transcrição ARNTL/genética
3.
Semin Immunopathol ; 44(2): 225-237, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35022891

RESUMO

It has emerged that an interconnected relationship exists between metabolism, circadian rhythms, and the immune system. The relationship between metabolism and circadian rhythms is not that surprising given the necessity to align rhythms of feeding/fasting with activity/rest. Recently, our understanding of the importance of metabolic pathways in terms of immune function, termed immunometabolism, has grown exponentially. It is now appreciated that the time of day during which the innate immune system is challenged strongly conditions the subsequent response. Recent observations have found that many individual components that make up the circadian clock also control aspects of metabolism in innate immune cells to modulate inflammation. This circadian/metabolic axis may be a key factor driving rhythmicity of immune function and circadian disruption is associated with a range of chronic inflammatory diseases such as atherosclerosis, obesity, and diabetes. The field of "circadian immunometabolism" seeks to reveal undiscovered circadian controlled metabolic pathways that in turn regulate immune responses. The innate immune system has been intricately linked to chronic inflammatory diseases, and within the immune system, individual cell types carry out unique roles in inflammation. Therefore, circadian immunometabolism effects are unique to each innate immune cell.


Assuntos
Relógios Circadianos , Imunidade Inata , Ritmo Circadiano , Humanos , Sistema Imunitário , Inflamação
4.
Nat Immunol ; 23(2): 153-154, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35079158

Assuntos
Ritmo Circadiano
5.
Front Immunol ; 12: 700431, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858390

RESUMO

The transcription factor BMAL1 is a clock protein that generates daily or circadian rhythms in physiological functions including the inflammatory response of macrophages. Intracellular metabolic pathways direct the macrophage inflammatory response, however whether the clock is impacting intracellular metabolism to direct this response is unclear. Specific metabolic reprogramming of macrophages controls the production of the potent pro-inflammatory cytokine IL-1ß. We now describe that the macrophage molecular clock, through Bmal1, regulates the uptake of glucose, its flux through glycolysis and the Krebs cycle, including the production of the metabolite succinate to drive Il-1ß production. We further demonstrate that BMAL1 modulates the level and localisation of the glycolytic enzyme PKM2, which in turn activates STAT3 to further drive Il-1ß mRNA expression. Overall, this work demonstrates that BMAL1 is a key metabolic sensor in macrophages, and its deficiency leads to a metabolic shift of enhanced glycolysis and mitochondrial respiration, leading to a heightened pro-inflammatory state. These data provide insight into the control of macrophage driven inflammation by the molecular clock, and the potential for time-based therapeutics against a range of chronic inflammatory diseases.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Inflamação/imunologia , Interleucina-1beta/metabolismo , Macrófagos/fisiologia , RNA Mensageiro/genética , Fatores de Transcrição ARNTL/genética , Animais , Relógios Circadianos , Glucose/metabolismo , Glicólise , Humanos , Interleucina-1beta/genética , Camundongos , Camundongos Knockout , Terapia de Alvo Molecular , Piruvato Quinase/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
6.
Nat Commun ; 11(1): 1055, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-32103022

RESUMO

Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma.


Assuntos
Asma/patologia , Caspases Iniciadoras/metabolismo , Dinoprostona/metabolismo , Macrófagos/imunologia , Piroptose/fisiologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Asma/imunologia , Caspases Iniciadoras/genética , Caspases Iniciadoras/imunologia , Células Cultivadas , Sinergismo Farmacológico , Feminino , Humanos , Indometacina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Misoprostol/farmacologia
7.
Trends Mol Med ; 25(7): 612-625, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31153819

RESUMO

Almost every cell has a molecular clock, which controls gene expression on a 24-h cycle, providing circadian rhythmicity. An example of a circadian behaviour common to most organisms is the feeding/fasting cycle, which shapes whole-body metabolism. However, the exact mechanisms by which the clock controls cellular metabolism have only recently become clear. The molecular clock and related metabolic pathways are also key drivers of immunity. Thus, a natural convergence of circadian biology, metabolism, and immunology has emerged to form a new field that we term 'circadian immunometabolism'. Expanding our understanding of this field will provide insights into chronic conditions such as obesity, cancer, diabetes, cardiovascular disease, and arthritis.


Assuntos
Relógios Biológicos/fisiologia , Metabolismo Energético , Imunomodulação , Animais , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Suscetibilidade a Doenças , Nível de Saúde , Homeostase , Humanos , Imunidade Inata , Mitocôndrias/metabolismo
8.
Nat Commun ; 10(1): 338, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30659183

RESUMO

Macrophages undergo metabolic changes during activation that are coupled to functional responses. The gram negative bacterial product lipopolysaccharide (LPS) is especially potent at driving metabolic reprogramming, enhancing glycolysis and altering the Krebs cycle. Here we describe a role for the citrate-derived metabolite malonyl-CoA in the effect of LPS in macrophages. Malonylation of a wide variety of proteins occurs in response to LPS. We focused on one of these, glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In resting macrophages, GAPDH binds to and suppresses translation of several inflammatory mRNAs, including that encoding TNFα. Upon LPS stimulation, GAPDH undergoes malonylation on lysine 213, leading to its dissociation from TNFα mRNA, promoting translation. We therefore identify for the first time malonylation as a signal, regulating GAPDH mRNA binding to promote inflammation.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Mediadores da Inflamação/farmacologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Animais , Citocinas/metabolismo , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Lisina/metabolismo , Malonil Coenzima A/metabolismo , Camundongos Endogâmicos C57BL , Mutagênese , Polirribossomos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(36): E8460-E8468, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30127006

RESUMO

A variety of innate immune responses and functions are dependent on time of day, and many inflammatory conditions are associated with dysfunctional molecular clocks within immune cells. However, the functional importance of these innate immune clocks has yet to be fully characterized. NRF2 plays a critical role in the innate immune system, limiting inflammation via reactive oxygen species (ROS) suppression and direct repression of the proinflammatory cytokines, IL-1ß and IL-6. Here we reveal that the core molecular clock protein, BMAL1, controls the mRNA expression of Nrf2 via direct E-box binding to its promoter to regulate its activity. Deletion of Bmal1 decreased the response of NRF2 to LPS challenge, resulting in a blunted antioxidant response and reduced synthesis of glutathione. ROS accumulation was increased in Bmal1-/- macrophages, facilitating accumulation of the hypoxic response protein, HIF-1α. Increased ROS and HIF-1α levels, as well as decreased activity of NRF2 in cells lacking BMAL1, resulted in increased production of the proinflammatory cytokine, IL-1ß. The excessive prooxidant and proinflammatory phenotype of Bmal1-/- macrophages was rescued by genetic and pharmacological activation of NRF2, or through addition of antioxidants. Our findings uncover a clear role for the molecular clock in regulating NRF2 in innate immune cells to control the inflammatory response. These findings provide insights into the pathology of inflammatory conditions, in which the molecular clock, oxidative stress, and IL-1ß are known to play a role.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fatores de Transcrição ARNTL/genética , Animais , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Interleucina-1beta/genética , Lipopolissacarídeos/toxicidade , Macrófagos/patologia , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio/metabolismo
10.
Nature ; 556(7699): 113-117, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29590092

RESUMO

The endogenous metabolite itaconate has recently emerged as a regulator of macrophage function, but its precise mechanism of action remains poorly understood. Here we show that itaconate is required for the activation of the anti-inflammatory transcription factor Nrf2 (also known as NFE2L2) by lipopolysaccharide in mouse and human macrophages. We find that itaconate directly modifies proteins via alkylation of cysteine residues. Itaconate alkylates cysteine residues 151, 257, 288, 273 and 297 on the protein KEAP1, enabling Nrf2 to increase the expression of downstream genes with anti-oxidant and anti-inflammatory capacities. The activation of Nrf2 is required for the anti-inflammatory action of itaconate. We describe the use of a new cell-permeable itaconate derivative, 4-octyl itaconate, which is protective against lipopolysaccharide-induced lethality in vivo and decreases cytokine production. We show that type I interferons boost the expression of Irg1 (also known as Acod1) and itaconate production. Furthermore, we find that itaconate production limits the type I interferon response, indicating a negative feedback loop that involves interferons and itaconate. Our findings demonstrate that itaconate is a crucial anti-inflammatory metabolite that acts via Nrf2 to limit inflammation and modulate type I interferons.


Assuntos
Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/química , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/agonistas , Fator 2 Relacionado a NF-E2/metabolismo , Succinatos/metabolismo , Alquilação , Animais , Carboxiliases , Bovinos , Cisteína/química , Cisteína/metabolismo , Citocinas/biossíntese , Citocinas/imunologia , Retroalimentação Fisiológica , Feminino , Células HEK293 , Humanos , Hidroliases/biossíntese , Interferon beta/imunologia , Interferon beta/farmacologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Proteínas/metabolismo , Ratos , Ratos Wistar , Succinatos/química
11.
J Biol Chem ; 293(15): 5509-5521, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29463677

RESUMO

Different immune activation states require distinct metabolic features and activities in immune cells. For instance, inhibition of fatty acid synthase (FASN), which catalyzes the synthesis of long-chain fatty acids, prevents the proinflammatory response in macrophages; however, the precise role of this enzyme in this response remains poorly defined. Consistent with previous studies, we found here that FASN is essential for lipopolysaccharide-induced, Toll-like receptor (TLR)-mediated macrophage activation. Interestingly, only agents that block FASN upstream of acetoacetyl-CoA synthesis, including the well-characterized FASN inhibitor C75, inhibited TLR4 signaling, while those acting downstream had no effect. We found that acetoacetyl-CoA could overcome C75's inhibitory effect, whereas other FASN metabolites, including palmitate, did not prevent C75-mediated inhibition. This suggested an unexpected role for acetoacetyl-CoA in inflammation that is independent of its role in palmitate synthesis. Our evidence further suggested that acetoacetyl-CoA arising from FASN activity promotes cholesterol production, indicating a surprising link between fatty acid synthesis and cholesterol synthesis. We further demonstrate that this process is required for TLR4 to enter lipid rafts and facilitate TLR4 signaling. In conclusion, we have uncovered an unexpected link between FASN and cholesterol synthesis that appears to be required for TLR signal transduction and proinflammatory macrophage activation.


Assuntos
Colesterol/biossíntese , Ácido Graxo Sintase Tipo I/metabolismo , Ativação de Macrófagos , Macrófagos/enzimologia , Transdução de Sinais , Acil Coenzima A/metabolismo , Animais , Inflamação/enzimologia , Camundongos , Ácido Palmítico/metabolismo , Receptor 4 Toll-Like/metabolismo
12.
Trends Mol Med ; 23(2): 165-180, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28109721

RESUMO

The inflammasome is a cytoplasmic protein complex that processes interleukins (IL)-1ß and IL-18, and drives a form of cell death known as pyroptosis. Oligomerization of this complex is actually the second step of activation, and a priming step must occur first. This involves transcriptional upregulation of pro-IL-1ß, inflammasome sensor NLRP3, or the non-canonical inflammasome sensor caspase-11. An additional aspect of priming is the post-translational modification of particular inflammasome constituents. Priming is typically accomplished in vitro using a microbial Toll-like receptor (TLR) ligand. However, it is now clear that inflammasomes are activated during the progression of sterile inflammatory diseases such as atherosclerosis, metabolic disease, and neuroinflammatory disorders. Therefore, it is time to consider the endogenous factors and mechanisms that may prime the inflammasome in these conditions.


Assuntos
Inflamassomos/imunologia , Inflamação/imunologia , Doença de Alzheimer/imunologia , Animais , Aterosclerose/imunologia , Doenças Hereditárias Autoinflamatórias/imunologia , Humanos , Imunidade Inata , Interleucina-1beta/imunologia , Doenças Metabólicas/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neuroimunomodulação , Obesidade/imunologia , Receptores Toll-Like/imunologia
13.
Cell Rep ; 9(4): 1538-53, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456142

RESUMO

Mitochondrial depolarization promotes Parkin- and PTEN-induced kinase 1 (PINK1)-dependent polyubiquitination of multiple proteins on mitochondrial outer membranes, resulting in the removal of defective mitochondria via mitophagy. Because Parkin mutations occur in Parkinson's disease, a condition associated with the death of dopaminergic neurons in the midbrain, wild-type Parkin is thought to promote neuronal survival. However, here we show that wild-type Parkin greatly sensitized toward apoptosis induced by mitochondrial depolarization but not by proapoptotic stimuli that failed to activate Parkin. Parkin-dependent apoptosis required PINK1 and was efficiently blocked by prosurvival members of the Bcl-2 family or knockdown of Bax and Bak. Upon mitochondrial depolarization, the Bcl-2 family member Mcl-1 underwent rapid Parkin- and PINK1-dependent polyubiquitination and degradation, which sensitized toward apoptosis via opening of the Bax/Bak channel. These data suggest that similar to other sensors of cell stress, such as p53, Parkin has cytoprotective (mitophagy) or cytotoxic modes (apoptosis), depending on the degree of mitochondrial damage.


Assuntos
Apoptose , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Poliubiquitina/metabolismo , Proteínas Quinases/metabolismo , Transporte Proteico , Ubiquitinação , Proteína X Associada a bcl-2/metabolismo
14.
Mol Cell ; 55(3): 451-66, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-24999239

RESUMO

Mitophagy facilitates the selective elimination of impaired or depolarized mitochondria through targeting the latter to autophagosomes. Parkin becomes localized to depolarized mitochondria in a PINK1-dependent manner and polyubiquitinates multiple mitochondrial outer membrane proteins. This permits ubiquitin-binding proteins (e.g., p62 and NBR1) to target impaired mitochondria to autophagosomes via Atg8/LC3II. Bcl-2 family proteins regulate mitochondrial outer membrane permeabilization during apoptosis and can also influence macroautophagy via interactions with Beclin-1. Here, we show that Parkin-dependent mitophagy is antagonized by prosurvival members of the Bcl-2 family (e.g., Bcl-xL and Mcl-1) in a Beclin-1-independent manner. Bcl-2 proteins suppressed mitophagy through inhibition of Parkin translocation to depolarized mitochondria. Consistent with this, Parkin translocation to mitochondria was enhanced by BH3-only proteins or a BH3-only mimetic. Taken together with their role as regulators of apoptosis-associated mitochondrial permeabilization, as well as mitochondrial fission/fusion dynamics, this suggests that Bcl-2 family proteins act as global regulators of mitochondrial homeostasis.


Assuntos
Mitocôndrias/fisiologia , Dinâmica Mitocondrial , Mitofagia , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Transdução de Sinais , Proteína bcl-X/metabolismo , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
Cancer Cell ; 23(4): 425-6, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23597558

RESUMO

Autophagy, a process for recycling cellular constituents, is normally associated with cell survival and is thought to be beneficial for tumor maintenance. However, in this issue of Cancer Cell, Lamy and colleagues report that multiple myeloma utilizes caspase-10 to restrain autophagy and undergoes autophagic cell death upon its removal or inhibition.

16.
Hum Gene Ther ; 24(3): 245-58, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23360514

RESUMO

Since HIV requires CD4 and a co-receptor, most commonly C-C chemokine receptor 5 (CCR5), for cellular entry, targeting CCR5 expression is an attractive approach for therapy of HIV infection. Treatment of CD4(+) T cells with zinc-finger protein nucleases (ZFNs) specifically disrupting chemokine receptor CCR5 coding sequences induces resistance to HIV infection in vitro and in vivo. A chimeric Ad5/F35 adenoviral vector encoding CCR5-ZFNs permitted efficient delivery and transient expression following anti-CD3/anti-CD28 costimulation of T lymphocytes. We present data showing CD3/CD28 costimulation substantially improved transduction efficiency over reported methods for Ad5/F35 transduction of T lymphocytes. Modifications to the laboratory scale process, incorporating clinically compatible reagents and methods, resulted in a robust ex vivo manufacturing process capable of generating >10(10) CCR5 gene-edited CD4+ T cells from healthy and HIV+ donors. CD4+ T-cell phenotype, cytokine production, and repertoire were comparable between ZFN-modified and control cells. Following consultation with regulatory authorities, we conducted in vivo toxicity studies that showed no detectable ZFN-specific toxicity or T-cell transformation. Based on these findings, we initiated a clinical trial testing the safety and feasibility of CCR5 gene-edited CD4+ T-cell transfer in study subjects with HIV-1 infection.


Assuntos
Enzimas de Restrição do DNA/genética , Vetores Genéticos/normas , Infecções por HIV/genética , Infecções por HIV/imunologia , Receptores CCR5/genética , Dedos de Zinco/genética , Adenovírus Humanos/genética , Transferência Adotiva , Animais , Antígenos CD28/imunologia , Complexo CD3/imunologia , Enzimas de Restrição do DNA/metabolismo , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Infecções por HIV/terapia , Humanos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Fenótipo , Receptores CCR5/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transdução Genética/métodos , Transdução Genética/normas , Transplante Heterólogo
17.
Hum Gene Ther ; 22(12): 1575-86, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21838572

RESUMO

Cytotoxic T lymphocytes (CTLs) modified with chimeric antigen receptors (CARs) for adoptive immunotherapy of hematologic malignancies are effective in preclinical models and are being tested in several clinical trials. Although CTLs bearing stably expressed CARs generated by integrating viral vectors are efficacious and have potential long-term persistence, this mechanism of CAR expression can potentially result in significant toxicity. T cells were electroporated with an optimized in vitro transcribed RNA encoding a CAR against CD19. These RNA CAR CTLs were then tested in vitro and in vivo for efficacy. We found that T cells expressing an anti-CD19 CAR introduced by electroporation with optimized mRNA were potent and specific killers of CD19 target cells. CD19 RNA CAR T cells given to immunodeficient mice bearing xenografted leukemia rapidly migrated to sites of disease and retained significant target-specific lytic activity. Unexpectedly, a single injection of CD19 RNA CAR T cells reduced disease burden within 1 day after administration, resulting in a significant prolongation of survival in an aggressive leukemia xenograft model. The surface expression of the RNA CARs may be titrated, giving T cells with potentially tunable levels of effector functions such as cytokine release and cytotoxicity. RNA CARs are a genetic engineering approach that should not be subject to genotoxicity, and they provide a platform for rapidly optimizing CAR design before proceeding to more costly and laborious stable expression systems.


Assuntos
Imunoterapia Adotiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , RNA Mensageiro/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T/transplante , Animais , Antígenos CD19/genética , Western Blotting , Células Cultivadas , Citotoxicidade Imunológica/imunologia , Eletroporação , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas Ligadas por GPI/genética , Humanos , Lentivirus/genética , Mesotelina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T/metabolismo , Carga Tumoral/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Am J Respir Crit Care Med ; 184(12): 1395-9, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21642245

RESUMO

New therapeutic strategies are needed for malignant pleural mesothelioma (MPM). We conducted a single-center, open-label, nonrandomized, pilot and feasibility trial using two intrapleural doses of an adenoviral vector encoding human IFN-α (Ad.IFN-α2b). Nine subjects were enrolled at two dose levels. The first three subjects had very high pleural and systemic IFN-α concentrations resulting in severe "flu-like" symptoms necessitating dose de-escalation. The next six patients had reduced (but still significant) pleural and serum IFN-α levels, but with tolerable symptoms. Repeated vector administration appeared to prolong IFN-α expression levels. Anti-tumor humoral immune responses against mesothelioma cell lines were seen in seven of the eight subjects evaluated. No clinical responses were seen in the four subjects with advanced disease. However, evidence of disease stability or tumor regression was seen in the remaining five patients, including one dramatic example of partial tumor regression at sites not in contiguity with vector infusion. These data show that Ad.IFN-α2b has potential therapeutic benefit in MPM and that it generates anti-tumor immune responses that may induce anatomic and/or metabolic reductions in distant tumor. Clinical trial registered with www.clinicaltrials.gov (NCT 01212367).


Assuntos
Terapia Genética , Fatores Imunológicos/administração & dosagem , Interferon-alfa/administração & dosagem , Mesotelioma/terapia , Neoplasias Pleurais/terapia , Adenoviridae , Idoso , Idoso de 80 Anos ou mais , Estudos de Viabilidade , Feminino , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Fatores Imunológicos/genética , Interferon alfa-2 , Interferon-alfa/genética , Masculino , Mesotelioma/diagnóstico por imagem , Mesotelioma/imunologia , Pessoa de Meia-Idade , Imagem Multimodal , Projetos Piloto , Neoplasias Pleurais/diagnóstico por imagem , Neoplasias Pleurais/imunologia , Tomografia por Emissão de Pósitrons , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Tomografia Computadorizada por Raios X
19.
Cancer Res ; 70(22): 9053-61, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20926399

RESUMO

Redirecting T lymphocyte antigen specificity by gene transfer can provide large numbers of tumor-reactive T lymphocytes for adoptive immunotherapy. However, safety concerns associated with viral vector production have limited clinical application of T cells expressing chimeric antigen receptors (CAR). T lymphocytes can be gene modified by RNA electroporation without integration-associated safety concerns. To establish a safe platform for adoptive immunotherapy, we first optimized the vector backbone for RNA in vitro transcription to achieve high-level transgene expression. CAR expression and function of RNA-electroporated T cells could be detected up to a week after electroporation. Multiple injections of RNA CAR-electroporated T cells mediated regression of large vascularized flank mesothelioma tumors in NOD/scid/γc(-/-) mice. Dramatic tumor reduction also occurred when the preexisting intraperitoneal human-derived tumors, which had been growing in vivo for >50 days, were treated by multiple injections of autologous human T cells electroporated with anti-mesothelin CAR mRNA. This is the first report using matched patient tumor and lymphocytes showing that autologous T cells from cancer patients can be engineered to provide an effective therapy for a disseminated tumor in a robust preclinical model. Multiple injections of RNA-engineered T cells are a novel approach for adoptive cell transfer, providing flexible platform for the treatment of cancer that may complement the use of retroviral and lentiviral engineered T cells. This approach may increase the therapeutic index of T cells engineered to express powerful activation domains without the associated safety concerns of integrating viral vectors.


Assuntos
Imunoterapia Adotiva/métodos , Mesotelioma/terapia , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antígenos CD19/genética , Antígenos CD19/metabolismo , Células Cultivadas , Citotoxicidade Imunológica/imunologia , Eletroporação , Citometria de Fluxo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Injeções , Células K562 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mesotelina , Mesotelioma/imunologia , Mesotelioma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/metabolismo , Carga Tumoral/imunologia , Células Tumorais Cultivadas
20.
Sci Transl Med ; 2(55): 55ra78, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-20980695

RESUMO

Human T helper 17 (T(H)17) cells regulate host defense, autoimmunity, and tumor immunity. Although cytokines that control human T(H)17 cell development have been identified, the costimulatory molecules important for T(H)17 cell generation are unknown. Here, we found that the inducible costimulator (ICOS) was critical for the differentiation and expansion of human T(H)17 cells. Human cord blood contained a subset of CD161(+)CD4(+) T cells that were recent emigrants from the thymus, expressed ICOS constitutively, and were imprinted as T(H)17 cells through ICOS signaling. ICOS stimulation induced c-MAF, RORC2, and T-bet expression in these cells, leading to increased secretion of interleukin-21 (IL-21), IL-17, and interferon-γ (IFN-γ) compared with cells stimulated with CD28. Conversely, CD28 ligation abrogated ICOS costimulation, dampening RORC2 expression while promoting the expression of the aryl hydrocarbon receptor, which led to reduced secretion of IL-17 and enhanced production of IL-22 compared with cells stimulated with ICOS. Moreover, ICOS promoted the robust expansion of IL-17(+)IFN-γ(+) human T cells, and the antitumor activity of these cells after adoptive transfer into mice bearing large human tumors was superior to that of cells expanded with CD28. The therapeutic effectiveness of ICOS-expanded cells was associated with enhanced functionality and engraftment in vivo. These findings reveal a vital role for ICOS signaling in the generation and maintenance of human T(H)17 cells and suggest that components of this pathway could be therapeutically targeted to treat cancer or chronic infection and, conversely, that interruption of this pathway may have utility in multiple sclerosis and other autoimmune syndromes. These findings have provided the rationale for designing new clinical trials for tumor immunotherapy.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Células Th17 , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos CD28/genética , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Sangue Fetal/citologia , Humanos , Imunoterapia/métodos , Proteína Coestimuladora de Linfócitos T Induzíveis , Interferon gama/genética , Interferon gama/imunologia , Interleucinas/genética , Interleucinas/imunologia , Camundongos , Subfamília B de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília B de Receptores Semelhantes a Lectina de Células NK/imunologia , Neoplasias/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Proteínas Proto-Oncogênicas c-maf/genética , Proteínas Proto-Oncogênicas c-maf/imunologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th17/imunologia , Células Th17/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA