Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 31(6): 1533-1549, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-36620962

RESUMO

RNA therapeutics have had a tremendous impact on medicine, recently exemplified by the rapid development and deployment of mRNA vaccines to combat the COVID-19 pandemic. In addition, RNA-targeting drugs have been developed for diseases with significant unmet medical needs through selective mRNA knockdown or modulation of pre-mRNA splicing. Recently, RNA editing, particularly antisense RNA-guided adenosine deaminase acting on RNA (ADAR)-based programmable A-to-I editing, has emerged as a powerful tool to manipulate RNA to enable correction of disease-causing mutations and modulate gene expression and protein function. Beyond correcting pathogenic mutations, the technology is particularly well suited for therapeutic applications that require a transient pharmacodynamic effect, such as the treatment of acute pain, obesity, viral infection, and inflammation, where it would be undesirable to introduce permanent alterations to the genome. Furthermore, transient modulation of protein function, such as altering the active sites of enzymes or the interface of protein-protein interactions, opens the door to therapeutic avenues ranging from regenerative medicine to oncology. These emerging RNA-editing-based toolsets are poised to broadly impact biotechnology and therapeutic applications. Here, we review the emerging field of therapeutic RNA editing, highlight recent laboratory advancements, and discuss the key challenges on the path to clinical development.


Assuntos
COVID-19 , RNA , Humanos , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Edição de RNA/genética , Pandemias , COVID-19/genética , COVID-19/terapia , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo
2.
Sci Adv ; 5(7): eaaw5096, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31281894

RESUMO

Chimeric antigen receptor (CAR) T cell therapy for hematologic malignancies is fraught with several unknowns, including number of functional T cells that engage target tumor, durability and subsequent expansion and contraction of that engagement, and whether toxicity can be managed. Non-invasive, serial imaging of CAR T cell therapy using a reporter transgene can address those issues quantitatively. We have transduced anti-CD19 CAR T cells with the prostate-specific membrane antigen (PSMA) because it is a human protein with restricted normal tissue expression and has an expanding array of positron emission tomography (PET) and therapeutic radioligands. We demonstrate that CD19-tPSMA(N9del) CAR T cells can be tracked with [18F]DCFPyL PET in a Nalm6 model of acute lymphoblastic leukemia. Divergence between the number of CD19-tPSMA(N9del) CAR T cells in peripheral blood and bone marrow and those in tumor was evident. These findings underscore the need for non-invasive repeatable monitoring of CAR T cell disposition clinically.


Assuntos
Antígenos de Superfície/metabolismo , Glutamato Carboxipeptidase II/metabolismo , Imunoterapia Adotiva , Tomografia por Emissão de Pósitrons/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico por imagem , Animais , Antígenos CD19/metabolismo , Antígenos de Superfície/genética , Glutamato Carboxipeptidase II/genética , Humanos , Leucemia Experimental/diagnóstico por imagem , Leucemia Experimental/patologia , Lisina/análogos & derivados , Camundongos Endogâmicos NOD , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/fisiologia , Ureia/análogos & derivados
3.
Front Cell Dev Biol ; 7: 35, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984757

RESUMO

During early avian development, primordial germ cells (PGC) are highly migratory, moving from the central area pellucida of the blastoderm to the anterior extra-embryonic germinal crescent. The PGCs soon move into the forming blood vessels by intravasation and travel in the circulatory system to the genital ridges where they participate in the organogenesis of the gonads. This complex cellular migration takes place in close association with a nascent extracellular matrix that matures in a precise spatio-temporal pattern. We first compiled a list of quail matrisome genes by bioinformatic screening of human matrisome orthologs. Next, we used single cell RNA-seq analysis (scRNAseq) to determine that PGCs express numerous ECM and ECM-associated genes in early embryos. The expression of select ECM transcripts and proteins in PGCs were verified by fluorescent in situ hybridization (FISH) and immunofluorescence (IF). Live imaging of transgenic quail embryos injected with fluorescent antibodies against fibronectin and laminin, showed that germinal crescent PGCs display rapid shape changes and morphological properties such as blebbing and filopodia while surrounded by, or in close contact with, an ECM fibril meshwork that is itself in constant motion. Injection of anti-ß1 integrin CSAT antibodies resulted in a reduction of mature fibronectin and laminin fibril meshwork in the germinal crescent at HH4-5 but did not alter the active motility of the PGCs or their ability to populate the germinal crescent. These results suggest that integrin ß1 receptors are important, but not required, for PGCs to successfully migrate during embryonic development, but instead play a vital role in ECM fibrillogenesis and assembly.

4.
J Neuroimmunol ; 294: 41-5, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27138097

RESUMO

Innate lymphoid cells (ILCs) play an important role in immunity, inflammation, and tissue remodeling and their dysregulation is implicated in autoimmune and inflammatory disorders. We analyzed the impact of daclizumab, a humanized monoclonal anti-CD25 antibody, on circulating natural killer (NK) cells and ILCs in a cohort of multiple sclerosis patients. An increase in CD56(bright) NK cells and CD56(hi)CD16(intermediate) transitional NK cells was observed. No significant change in total ILCs or major ILC subpopulations was observed. These results refine our understanding of the impact of daclizumab on innate lymphoid cell populations.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Imunoglobulina G/uso terapêutico , Imunossupressores/uso terapêutico , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD/metabolismo , Estudos de Coortes , Daclizumabe , Feminino , Citometria de Fluxo , Humanos , Imunoglobulina G/farmacologia , Imunossupressores/farmacologia , Linfócitos/metabolismo , Masculino , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia
5.
Immunology ; 148(3): 276-86, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27012310

RESUMO

Interleukin-2 (IL-2) is a critical regulator of immune homeostasis through its non-redundant role in regulatory T (Treg) cell biology. There is major interest in therapeutic modulation of the IL-2 pathway to promote immune activation in the context of tumour immunotherapy or to enhance immune suppression in the context of transplantation, autoimmunity and inflammatory diseases. Antibody-mediated targeting of the high-affinity IL-2 receptor α chain (IL-2Rα or CD25) offers a direct mechanism to target IL-2 biology and is being actively explored in the clinic. In mouse models, the rat anti-mouse CD25 clone PC61 has been used extensively to investigate the biology of IL-2 and Treg cells; however, there has been controversy and conflicting data on the exact in vivo mechanistic function of PC61. Engineering antibodies to alter Fc/Fc receptor interactions can significantly alter their in vivo function. In this study, we re-engineered the heavy chain constant region of an anti-CD25 monoclonal antibody to generate variants with highly divergent Fc effector function. Using these anti-CD25 Fc variants in multiple mouse models, we investigated the in vivo impact of CD25 blockade versus depletion of CD25(+) Treg cells on immune homeostasis. We report that immune homeostasis can be maintained during CD25 blockade but aberrant T-cell activation prevails when CD25(+) Treg cells are actively depleted. These results clarify the impact of PC61 on Treg cell biology and reveal an important distinction between CD25 blockade and depletion of CD25(+) Treg cells. These findings should inform therapeutic manipulation of the IL-2 pathway by targeting the high-affinity IL-2R.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunoglobulina G/farmacologia , Imunoterapia , Interleucina-2/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Anticorpos Monoclonais/genética , Anticorpos Antivirais/imunologia , Autoimunidade/imunologia , Fatores de Transcrição Forkhead/metabolismo , Homeostase/efeitos dos fármacos , Imunoglobulina G/genética , Terapia de Imunossupressão , Interleucina-2/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Engenharia de Proteínas , Ratos , Receptores de IgG/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T Reguladores/imunologia
6.
J Neuroimmunol ; 283: 74-85, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26004161

RESUMO

Fumarate-containing pharmaceuticals are potent therapeutic agents that influence multiple cellular pathways. Despite proven clinical efficacy, there is a significant lack of data that directly defines the molecular mechanisms of action of related, yet distinct fumarate compounds. We systematically compared the impact of dimethyl fumarate (DMF), monomethyl fumarate (MMF) and a mixture of monoethyl fumarate salts (Ca(++), Mg(++), Zn(++); MEF) on defined cellular responses. We demonstrate that DMF inhibited NF-κB-driven cytokine production and nuclear translocation of p65 and p52 in an Nrf2-independent manner. Equivalent doses of MMF and MEF did not affect NF-κB signaling. These results highlight a key difference in the biological impact of related, yet distinct fumarate compounds.


Assuntos
Fumaratos/farmacologia , NF-kappa B/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Neoplasias Ósseas/patologia , Linfoma de Burkitt/patologia , Cátions/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Fumarato de Dimetilo , Humanos , Técnicas In Vitro , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Maleatos/farmacologia , Camundongos , Camundongos Knockout , Estrutura Molecular , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/fisiologia , Subunidade p52 de NF-kappa B/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Osteossarcoma/patologia , Transdução de Sinais/efeitos dos fármacos , Baço/citologia , Fator de Transcrição RelA/metabolismo
7.
J Immunol ; 194(1): 84-92, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25416807

RESUMO

Regulatory T cells (Tregs) mediate immune tolerance to self and depend on IL-2 for homeostasis. Treg deficiency, dysfunction, and instability are implicated in the pathogenesis of numerous autoimmune diseases. There is considerable interest in therapeutic modulation of the IL-2 pathway to treat autoimmunity, facilitate transplantation tolerance, or potentiate tumor immunotherapy. Daclizumab is a humanized mAb that binds the IL-2 receptor a subunit (IL-2R a or CD25) and prevents IL-2 binding. In this study, we investigated the effect of daclizumab-mediated CD25 blockade on Treg homeostasis in patients with relapsing-remitting multiple sclerosis. We report that daclizumab therapy caused an ~50% decrease in Tregs over a 52-wk period. Remaining FOXP3+ cells retained a demethylated Treg-specific demethylated region in the FOXP3 promoter, maintained active cell cycling, and had minimal production of IL-2, IFN- g, and IL-17. In the presence of daclizumab, IL-2 serum concentrations increased and IL-2R bg signaling induced STAT5 phosphorylation and sustained FOXP3 expression. Treg declines were not associated with daclizumab-related clinical benefit or cutaneous adverse events. These results demonstrate that Treg phenotype and lineage stability can be maintained in the face of CD25 blockade.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Imunoglobulina G/uso terapêutico , Imunossupressores/uso terapêutico , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Interleucina-2/imunologia , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Linfócitos T Reguladores/efeitos dos fármacos , Contagem de Linfócito CD4 , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Daclizumabe , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Subunidade gama Comum de Receptores de Interleucina/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Interleucina-2/biossíntese , Interleucina-2/sangue , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Fosforilação , Regiões Promotoras Genéticas , Fator de Transcrição STAT5/metabolismo , Tolerância a Antígenos Próprios/efeitos dos fármacos , Tolerância a Antígenos Próprios/imunologia , Linfócitos T Reguladores/imunologia
8.
J Biol Chem ; 287(33): 28017-26, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22733812

RESUMO

Dimethyl fumarate (DMF) is an effective novel treatment for multiple sclerosis in clinical trials. A reduction of IFN-γ-producing CD4(+) T cells is observed in DMF-treated patients and may contribute to its clinical efficacy. However, the cellular and molecular mechanisms behind this clinical observation are unclear. In this study, we investigated the effects of DMF on dendritic cell (DC) maturation and subsequent DC-mediated T cell responses. We show that DMF inhibits DC maturation by reducing inflammatory cytokine production (IL-12 and IL-6) and the expression of MHC class II, CD80, and CD86. Importantly, this immature DC phenotype generated fewer activated T cells that were characterized by decreased IFN-γ and IL-17 production. Further molecular studies demonstrated that DMF impaired nuclear factor κB (NF-κB) signaling via reduced p65 nuclear translocalization and phosphorylation. NF-κB signaling was further decreased by DMF-mediated suppression of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and its downstream kinase mitogen stress-activated kinase 1 (MSK1). MSK1 suppression resulted in decreased p65 phosphorylation at serine 276 and reduced histone phosphorylation at serine 10. As a consequence, DMF appears to reduce p65 transcriptional activity both directly and indirectly by promoting a silent chromatin environment. Finally, treatment of DCs with the MSK1 inhibitor H89 partially mimicked the effects of DMF on the DC signaling pathway and impaired DC maturation. Taken together, these studies indicate that by suppression of both NF-κB and ERK1/2-MSK1 signaling, DMF inhibits maturation of DCs and subsequently Th1 and Th17 cell differentiation.


Assuntos
Células Dendríticas/imunologia , Fumaratos/farmacologia , Imunossupressores/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteínas Quinases S6 Ribossômicas 90-kDa/imunologia , Fator de Transcrição RelA/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-1/imunologia , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Fumarato de Dimetilo , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/imunologia , Interleucina-12/imunologia , Interleucina-12/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo , Fator de Transcrição RelA/metabolismo
9.
Eur J Immunol ; 41(10): 2987-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21728174

RESUMO

Effector Th1 cells perpetuate inflammatory damage in a number of autoimmune diseases, including MS and its animal model EAE. Recently, a self-regulatory mechanism was described in which effector Th1 cells produce the immunomodulatory cytokine IL-10 to dampen the inflammatory response in both normal and autoimmune inflammation. While the presence of TGF-ß has been suggested to enhance and stabilize an IFN-γ(+) IL-10(+) phenotype, the molecular mechanism is poorly understood. Additionally, in the context of adoptive transfer EAE, it is unclear whether IL-10 acts on the transferred Th1 cells or on endogenous host cells. In the present study, using myelin-specific TCR-Tg mice, we show that repetitive Ag stimulation of effector Th1 cells in the presence of TGF-ß increases the population of IFN-γ(+) IL-10(+) cells, which correlates with a decrease in EAE severity. Additionally, TGF-ß signaling causes binding of Smad4 to the IL-10 promoter, providing molecular evidence for TGF-ß-mediated IL-10 production from Th1 effector cells. Finally, this study demonstrates that IL-10 not only reduces encephalitogenic markers such as IFN-γ and T-bet on Th1 effector cells expressing the IL-10R but also prevents recruitment of both transferred and host-derived inflammatory T cells. These data establish a regulatory mechanism by which highly activated Th1 effector cells modulate their pathogenicity through the induction of IL-10.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Interleucina-10/biossíntese , Proteína Smad4/metabolismo , Células Th1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transferência Adotiva , Animais , Células Cultivadas , Encefalomielite Autoimune Experimental/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/biossíntese , Interferon gama/biossíntese , Interleucina-10/genética , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Proteínas com Domínio T/biossíntese , Células Th1/imunologia
11.
J Immunol ; 184(10): 5628-36, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20393141

RESUMO

Myelin-specific effector Th1 cells are able to perpetuate CNS inflammation in experimental autoimmune encephalomyelitis, an animal model representative of multiple sclerosis. Although the effects of cytokines in the CNS microenvironment on naive CD4(+) T cells have been well described, much less is known about their ability to influence Ag-experienced effector cells. TGF-beta is a multifunctioning cytokine present in the healthy and inflamed CNS with well-characterized suppressive effects on naive T cell functions. However, the effects of TGF-beta on effector Th1 cells are not well defined. Using myelin-specific TCR transgenic mice, we demonstrate that TGF-beta elicits differential effects on naive versus effector Th1 cells. TGF-beta enhances cellular activation, proliferation, and cytokine production of effector Th1 cells; however, adoptive transfer of these cells into naive mice showed a reduction in encephalitogenicity. We subsequently demonstrate that the reduced encephalitogenic capacity is due to the ability of TGF-beta to promote the self-regulation of Th1 effector cells via IL-10 production. These data demonstrate a mechanism by which TGF-beta is able to suppress the encephalitogenicity of myelin-specific Th1 effector cells that is unique from its suppression of naive T cells.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Interleucina-10/fisiologia , Ativação Linfocitária/imunologia , Células Th1/imunologia , Fator de Crescimento Transformador beta1/fisiologia , Regulação para Cima/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/fisiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/prevenção & controle , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/fisiologia , Mediadores da Inflamação/fisiologia , Interleucina-10/antagonistas & inibidores , Interleucina-10/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Células Th1/metabolismo , Células Th1/transplante
12.
J Exp Med ; 206(7): 1549-64, 2009 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-19546248

RESUMO

The extent to which myelin-specific Th1 and Th17 cells contribute to the pathogenesis of experimental autoimmune encephalomyelitis (EAE) is controversial. Combinations of interleukin (IL)-1beta, IL-6, and IL-23 with transforming growth factor beta were used to differentiate myelin-specific T cell receptor transgenic T cells into Th17 cells, none of which could induce EAE, whereas Th1 cells consistently transferred disease. However, IL-6 was found to promote the differentiation of encephalitogenic Th17 cells. Further analysis of myelin-specific T cells that were encephalitogenic in spontaneous EAE and actively induced EAE demonstrated that T-bet expression was critical for pathogenicity, regardless of cytokine expression by the encephalitogenic T cells. These data suggest that encephalitogenicity of myelin-specific T cells appears to be mediated by a pathway dependent on T-bet and not necessarily pathway-specific end products, such as interferon gamma and IL-17.


Assuntos
Encefalomielite Autoimune Experimental , Interleucina-17/imunologia , Proteínas com Domínio T/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Animais , Diferenciação Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Interferon gama/imunologia , Interleucina-1beta/imunologia , Interleucina-23/imunologia , Interleucina-6/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Baço/citologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/fisiologia , Células Th1/citologia , Células Th1/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...