Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(9): e2216697120, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36802421

RESUMO

Peptide-binding proteins play key roles in biology, and predicting their binding specificity is a long-standing challenge. While considerable protein structural information is available, the most successful current methods use sequence information alone, in part because it has been a challenge to model the subtle structural changes accompanying sequence substitutions. Protein structure prediction networks such as AlphaFold model sequence-structure relationships very accurately, and we reasoned that if it were possible to specifically train such networks on binding data, more generalizable models could be created. We show that placing a classifier on top of the AlphaFold network and fine-tuning the combined network parameters for both classification and structure prediction accuracy leads to a model with strong generalizable performance on a wide range of Class I and Class II peptide-MHC interactions that approaches the overall performance of the state-of-the-art NetMHCpan sequence-based method. The peptide-MHC optimized model shows excellent performance in distinguishing binding and non-binding peptides to SH3 and PDZ domains. This ability to generalize well beyond the training set far exceeds that of sequence-only models and should be particularly powerful for systems where less experimental data are available.


Assuntos
Antígenos de Histocompatibilidade Classe II , Peptídeos , Ligação Proteica , Peptídeos/química , Antígenos de Histocompatibilidade Classe II/metabolismo , Genes MHC da Classe II , Domínios PDZ
2.
Nat Biotechnol ; 41(7): 919-931, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36593411

RESUMO

Ultrasound allows imaging at a much greater depth than optical methods, but existing genetically encoded acoustic reporters for in vivo cellular imaging have been limited by poor sensitivity, specificity and in vivo expression. Here we describe two acoustic reporter genes (ARGs)-one for use in bacteria and one for use in mammalian cells-identified through a phylogenetic screen of candidate gas vesicle gene clusters from diverse bacteria and archaea that provide stronger ultrasound contrast, produce non-linear signals distinguishable from background tissue and have stable long-term expression. Compared to their first-generation counterparts, these improved bacterial and mammalian ARGs produce 9-fold and 38-fold stronger non-linear contrast, respectively. Using these new ARGs, we non-invasively imaged in situ tumor colonization and gene expression in tumor-homing therapeutic bacteria, tracked the progression of tumor gene expression and growth in a mouse model of breast cancer, and performed gene-expression-guided needle biopsies of a genetically mosaic tumor, demonstrating non-invasive access to dynamic biological processes at centimeter depth.


Assuntos
Neoplasias , Animais , Camundongos , Genes Reporter/genética , Filogenia , Neoplasias/genética , Neoplasias/terapia , Bactérias/genética , Acústica , Mamíferos
3.
Nat Commun ; 13(1): 1585, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35332124

RESUMO

Rapid advances in synthetic biology are driving the development of genetically engineered microbes as therapeutic agents for a multitude of human diseases, including cancer. The immunosuppressive microenvironment of solid tumors, in particular, creates a favorable niche for systemically administered bacteria to engraft and release therapeutic payloads. However, such payloads can be harmful if released outside the tumor in healthy tissues where the bacteria also engraft in smaller numbers. To address this limitation, we engineer therapeutic bacteria to be controlled by focused ultrasound, a form of energy that can be applied noninvasively to specific anatomical sites such as solid tumors. This control is provided by a temperature-actuated genetic state switch that produces lasting therapeutic output in response to briefly applied focused ultrasound hyperthermia. Using a combination of rational design and high-throughput screening we optimize the switching circuits of engineered cells and connect their activity to the release of immune checkpoint inhibitors. In a clinically relevant cancer model, ultrasound-activated therapeutic microbes successfully turn on in situ and induce a marked suppression of tumor growth. This technology provides a critical tool for the spatiotemporal targeting of potent bacterial therapeutics in a variety of biological and clinical scenarios.


Assuntos
Imunoterapia , Neoplasias , Bactérias/genética , Engenharia Genética , Humanos , Neoplasias/terapia , Biologia Sintética , Microambiente Tumoral
4.
Nat Nanotechnol ; 16(12): 1403-1412, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34580468

RESUMO

Recent advances in molecular engineering and synthetic biology provide biomolecular and cell-based therapies with a high degree of molecular specificity, but limited spatiotemporal control. Here we show that biomolecules and cells can be engineered to deliver potent mechanical effects at specific locations inside the body through ultrasound-induced inertial cavitation. This capability is enabled by gas vesicles, a unique class of genetically encodable air-filled protein nanostructures. We show that low-frequency ultrasound can convert these biomolecules into micrometre-scale cavitating bubbles, unleashing strong local mechanical effects. This enables engineered gas vesicles to serve as remotely actuated cell-killing and tissue-disrupting agents, and allows genetically engineered cells to lyse, release molecular payloads and produce local mechanical damage on command. We demonstrate the capabilities of biomolecular inertial cavitation in vitro, in cellulo and in vivo, including in a mouse model of tumour-homing probiotic therapy.


Assuntos
Acústica , Gases/química , Técnicas Genéticas , Microbolhas , Animais , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Feminino , Humanos , Imunoterapia , Camundongos Endogâmicos BALB C , Imagem Óptica , Probióticos/farmacologia , Receptores de Superfície Celular/metabolismo , Ultrassonografia
5.
ACS Synth Biol ; 9(8): 1941-1950, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32786924

RESUMO

Genetically engineered T-cells are being developed to perform a variety of therapeutic functions. However, no robust mechanisms exist to externally control the activity of T-cells at specific locations within the body. Such spatiotemporal control could help mitigate potential off-target toxicity due to incomplete molecular specificity in applications such as T-cell immunotherapy against solid tumors. Temperature is a versatile external control signal that can be delivered to target tissues in vivo using techniques such as focused ultrasound and magnetic hyperthermia. Here, we test the ability of heat shock promoters to mediate thermal actuation of genetic circuits in primary human T-cells in the well-tolerated temperature range of 37-42 °C, and introduce genetic architectures enabling the tuning of the amplitude and duration of thermal activation. We demonstrate the use of these circuits to control the expression of chimeric antigen receptors and cytokines, and the killing of target tumor cells. This technology provides a critical tool to direct the activity of T-cells after they are deployed inside the body.


Assuntos
Linfócitos T/metabolismo , Células Cultivadas , Citocinas/metabolismo , Engenharia Genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Regiões Promotoras Genéticas , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/citologia , Temperatura
6.
Nat Chem Biol ; 13(1): 75-80, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27842069

RESUMO

Temperature is a unique input signal that could be used by engineered microbial therapeutics to sense and respond to host conditions or spatially targeted external triggers such as focused ultrasound. To enable these possibilities, we present two families of tunable, orthogonal, temperature-dependent transcriptional repressors providing switch-like control of bacterial gene expression at thresholds spanning the biomedically relevant range of 32-46 °C. We integrate these molecular bioswitches into thermal logic circuits and demonstrate their utility in three in vivo microbial therapy scenarios, including spatially precise activation using focused ultrasound, modulation of activity in response to a host fever, and self-destruction after fecal elimination to prevent environmental escape. This technology provides a critical capability for coupling endogenous or applied thermal signals to cellular function in basic research, biomedical and industrial applications.


Assuntos
Antibacterianos/metabolismo , Escherichia coli/genética , Fezes/microbiologia , Febre , Regulação Bacteriana da Expressão Gênica , Proteínas Repressoras/metabolismo , Temperatura , Ultrassom , Animais , Antibacterianos/química , Escherichia coli/isolamento & purificação , Feminino , Camundongos , Viabilidade Microbiana , Proteínas Repressoras/química , Dermatopatias/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA