Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(38): e2207525119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36095208

RESUMO

Progress in bottom-up synthetic biology has stimulated the development of synthetic cells (SCs), autonomous protein-manufacturing particles, as dynamic biomimetics for replacing diseased natural cells and addressing medical needs. Here, we report that SCs genetically encoded to produce proangiogenic factors triggered the physiological process of neovascularization in mice. The SCs were constructed of giant lipid vesicles and were optimized to facilitate enhanced protein production. When introduced with the appropriate genetic code, the SCs synthesized a recombinant human basic fibroblast growth factor (bFGF), reaching expression levels of up to 9⋅106 protein copies per SC. In culture, the SCs induced endothelial cell proliferation, migration, tube formation, and angiogenesis-related intracellular signaling, confirming their proangiogenic activity. Integrating the SCs with bioengineered constructs bearing endothelial cells promoted the remodeling of mature vascular networks, supported by a collagen-IV basement membrane-like matrix. In vivo, prolonged local administration of the SCs in mice triggered the infiltration of blood vessels into implanted Matrigel plugs without recorded systemic immunogenicity. These findings emphasize the potential of SCs as therapeutic platforms for activating physiological processes by autonomously producing biological drugs inside the body.


Assuntos
Células Artificiais , Fatores de Crescimento de Fibroblastos , Neovascularização Fisiológica , Animais , Células Artificiais/transplante , Movimento Celular , Proliferação de Células , Colágeno Tipo IV/metabolismo , Células Endoteliais/fisiologia , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Humanos , Camundongos , Biossíntese de Proteínas
2.
Nanotechnology ; 28(43): 43LT01, 2017 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-28872058

RESUMO

Despite advances in cancer therapy, treating cancer after it has metastasized remains an unmet clinical challenge. In this study we demonstrate that 100 nm liposomes target triple-negative murine breast-cancer metastases post intravenous administration. Metastatic breast cancer was induced in BALB/c mice either experimentally, by a tail vein injection of 4T1 cells, or spontaneously, after implanting a primary tumor xenograft. To track their biodistribution in vivo the liposomes were labeled with multi-modal diagnostic agents, including indocyanine green and rhodamine for whole-animal fluorescent imaging, gadolinium for magnetic resonance imaging (MRI), and europium for a quantitative biodistribution analysis. The accumulation of liposomes in the metastases peaked at 24 h post the intravenous administration, similar to the time they peaked in the primary tumor. The efficiency of liposomal targeting to the metastatic tissue exceeded that of a non-liposomal agent by 4.5-fold. Liposomes were detected at very early stages in the metastatic progression, including metastatic lesions smaller than 2 mm in diameter. Surprisingly, while nanoparticles target breast cancer metastasis, they may also be found in elevated levels in the pre-metastatic niche, several days before metastases are visualized by MRI or histologically in the tissue. This study highlights the promise of diagnostic and therapeutic nanoparticles for treating metastatic cancer, possibly even for preventing the onset of the metastatic dissemination by targeting the pre-metastatic niche.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Sistemas de Liberação de Medicamentos/métodos , Lipossomos/farmacocinética , Neoplasias Pulmonares/diagnóstico por imagem , Metástase Neoplásica/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Linhagem Celular Tumoral , Európio/química , Európio/farmacocinética , Feminino , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Lipossomos/síntese química , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Metástase Neoplásica/patologia , Transplante de Neoplasias , Imagem Óptica , Rodaminas/química , Rodaminas/farmacocinética , Distribuição Tecidual , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/secundário
3.
Adv Mater ; 35(51): e2304654, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37753928

RESUMO

Monoclonal antibodies (mAbs) hold promise in treating Parkinson's disease (PD), although poor delivery to the brain hinders their therapeutic application. In the current study, it is demonstrated that brain-targeted liposomes (BTL) enhance the delivery of mAbs across the blood-brain-barrier (BBB) and into neurons, thereby allowing the intracellular and extracellular treatment of the PD brain. BTL are decorated with transferrin to improve brain targeting through overexpressed transferrin-receptors on the BBB during PD. BTL are loaded with SynO4, a mAb that inhibits alpha-synuclein (AS) aggregation, a pathological hallmark of PD. It is shown that 100-nm BTL cross human BBB models intact and are taken up by primary neurons. Within neurons, SynO4 is released from the nanoparticles and bound to its target, thereby reducing AS aggregation, and enhancing neuronal viability. In vivo, intravenous BTL administration results in a sevenfold increase in mAbs in brain cells, decreasing AS aggregation and neuroinflammation. Treatment with BTL also improve behavioral motor function and learning ability in mice, with a favorable safety profile. Accordingly, targeted nanotechnologies offer a valuable platform for drug delivery to treat brain neurodegeneration.


Assuntos
Doença de Parkinson , Animais , Humanos , Camundongos , alfa-Sinucleína/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Sintomas Comportamentais , Encéfalo/metabolismo , Lipossomos/metabolismo , Doença de Parkinson/tratamento farmacológico , Transferrinas
4.
Nat Commun ; 13(1): 2328, 2022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35484097

RESUMO

Development of regulated cellular processes and signaling methods in synthetic cells is essential for their integration with living materials. Light is an attractive tool to achieve this, but the limited penetration depth into tissue of visible light restricts its usability for in-vivo applications. Here, we describe the design and implementation of bioluminescent intercellular and intracellular signaling mechanisms in synthetic cells, dismissing the need for an external light source. First, we engineer light generating SCs with an optimized lipid membrane and internal composition, to maximize luciferase expression levels and enable high-intensity emission. Next, we show these cells' capacity to trigger bioprocesses in natural cells by initiating asexual sporulation of dark-grown mycelial cells of the fungus Trichoderma atroviride. Finally, we demonstrate regulated transcription and membrane recruitment in synthetic cells using bioluminescent intracellular signaling with self-activating fusion proteins. These functionalities pave the way for deploying synthetic cells as embeddable microscale light sources that are capable of controlling engineered processes inside tissues.


Assuntos
Células Artificiais , Optogenética , Luz , Luciferases , Optogenética/métodos , Transdução de Sinais
5.
J Vis Exp ; (158)2020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32391815

RESUMO

The bottom-up assembly approach for construction of synthetic cells is an effective tool for isolating and investigating cellular processes in a cell mimicking environment. Furthermore, the development of cell-free expression systems has demonstrated the ability to reconstitute the protein production, transcription and translation processes (DNA→RNA→protein) in a controlled manner, harnessing synthetic biology. Here we describe a protocol for preparing a cell-free expression system, including the production of a potent bacterial lysate and encapsulating this lysate inside cholesterol-rich lipid-based giant unilamellar vesicles (GUVs) (i.e., stable liposomes), to form synthetic cells. The protocol describes the methods for preparing the components of the synthetic cells including the production of active bacterial lysates, followed by a detailed step-by-step preparation of the synthetic cells based on a water-in-oil emulsion transfer method. These facilitate the production of millions of synthetic cells in a simple and affordable manner with a high versatility for producing different types of proteins. The obtained synthetic cells can be used to investigate protein/RNA production and activity in an isolated environment, in directed evolution, and also as a controlled drug delivery platform for on-demand production of therapeutic proteins inside the body.


Assuntos
Células Artificiais/metabolismo , Emulsões/química , Escherichia coli/metabolismo , Biossíntese de Proteínas , Biologia Sintética/métodos , Sistema Livre de Células/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Lipossomos/química , Luciferases/metabolismo
6.
J Control Release ; 296: 1-13, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30615983

RESUMO

Acidic pH in the tumor microenvironment is associated with cancer metabolism and creates a physiological barrier that prevents from drugs to penetrate cells. Specifically, ionizable weak-base drugs, such as doxorubicin, freely permeate membranes in their uncharged form, however, in the acidic tumor microenvironment these drugs become charged and their cellular permeability is retarded. In this study, 100-nm liposomes loaded with sodium bicarbonate were used as adjuvants to elevate the tumor pH. Combined treatment of triple-negative breast cancer cells (4T1) with doxorubicin and sodium-bicarbonate enhanced drug uptake and increased its anti-cancer activity. In vivo, mice bearing orthotropic 4T1 breast cancer tumors were administered either liposomal or free bicarbonate intravenously. 3.7 ±â€¯0.3% of the injected liposomal dose was detected in the tumor after twenty-four hours, compared to 0.17% ±â€¯0.04% in the group injected free non-liposomal bicarbonate, a 21-fold increase. Analyzing nanoparticle biodistribution within the tumor tissue revealed that 93% of the PEGylated liposomes accumulated in the extracellular matrix, while 7% were detected intracellularly. Mice administered bicarbonate-loaded liposomes reached an intra-tumor pH value of 7.38 ±â€¯0.04. Treating tumors with liposomal bicarbonate combined with a sub-therapeutic dose of doxorubicin achieved an improved therapeutic outcome, compared to mice treated with doxorubicin or bicarbonate alone. Interestingly, analysis of the tumor microenvironment demonstrated an increase in immune cell' population (T-cell, B-cell and macrophages) in tumors treated with liposomal bicarbonate. This study demonstrates that targeting metabolic adjuvants with nanoparticles to the tumor microenvironment can enhance anticancer drug activity and improve treatment.


Assuntos
Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias , Bicarbonato de Sódio/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Transporte Biológico/efeitos dos fármacos , Contagem de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacocinética , Feminino , Humanos , Concentração de Íons de Hidrogênio , Lipossomos , Camundongos Endogâmicos BALB C , Neoplasias/química , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Bicarbonato de Sódio/farmacocinética , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
7.
ACS Nano ; 13(10): 11008-11021, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31503443

RESUMO

Overexpressed extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) limits drug penetration into the tumor and is associated with poor prognosis. Here, we demonstrate that a pretreatment based on a proteolytic-enzyme nanoparticle system disassembles the dense PDAC collagen stroma and increases drug penetration into the pancreatic tumor. More specifically, the collagozome, a 100 nm liposome encapsulating collagenase, was rationally designed to protect the collagenase from premature deactivation and prolonged its release rate at the target site. Collagen is the main component of the PDAC stroma, reaching 12.8 ± 2.3% vol in diseased mice pancreases, compared to 1.4 ± 0.4% in healthy mice. Upon intravenous injection of the collagozome, ∼1% of the injected dose reached the pancreas over 8 h, reducing the level of fibrotic tissue to 5.6 ± 0.8%. The collagozome pretreatment allowed increased drug penetration into the pancreas and improved PDAC treatment. PDAC tumors, pretreated with the collagozome followed by paclitaxel micelles, were 87% smaller than tumors pretreated with empty liposomes followed by paclitaxel micelles. Interestingly, degrading the ECM did not increase the number of circulating tumor cells or metastasis. This strategy holds promise for degrading the extracellular stroma in other diseases as well, such as liver fibrosis, enhancing tissue permeability before drug administration.


Assuntos
Adenocarcinoma/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Colagenases/farmacologia , Nanopartículas/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Colágeno/química , Colágeno/genética , Colagenases/química , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Fibrose/tratamento farmacológico , Fibrose/patologia , Fibrose/prevenção & controle , Humanos , Lipossomos/química , Lipossomos/farmacologia , Camundongos , Nanopartículas/uso terapêutico , Paclitaxel/química , Paclitaxel/farmacologia , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Microambiente Tumoral/efeitos dos fármacos
8.
Sci Rep ; 8(1): 7589, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773873

RESUMO

As the world population grows, there is a need for efficient agricultural technologies to provide global food requirements and reduce environmental toll. In medicine, nanoscale drug delivery systems grant improved therapeutic precision by overcoming biological barriers and enhancing drug targeting to diseased tissues. Here, we loaded nanoscale drug-delivery systems with agricultural nutrients, and applied them to the leaves of tomato plants. We show that the nanoparticles - liposomes composed of plant-derived lipids, penetrate the leaf and translocate in a bidirectional manner, distributing to other leaves and to the roots. The liposomes were then internalized by the plant cells, where they released their active ingredient. Up to 33% of the applied nanoparticles penetrated the leaf, compared to less than one percent of free-molecules applied in a similar manner. In our study, tomato plants treated with liposomes loaded with Fe and Mg overcame acute nutrient deficiency which was not treatable using ordinary agricultural nutrients. Furthermore, to address regulatory concerns regarding airborne nanoparticles, we rationally designed liposomes that were stable only over short spraying distances (less than 2 meters), while the liposomes disintegrated into safe molecular building blocks (phospholipids) over longer airborne distances. These findings support expanding the implementation of nanotechnology for delivering micronutrients to agricultural crops for increasing yield.


Assuntos
Produtos Agrícolas/metabolismo , Sistemas de Liberação de Medicamentos , Lipossomos/química , Nanopartículas/administração & dosagem , Nutrientes/administração & dosagem , Folhas de Planta/metabolismo , Solanum lycopersicum/metabolismo , Produtos Agrícolas/crescimento & desenvolvimento , Solanum lycopersicum/crescimento & desenvolvimento , Nanopartículas/química , Folhas de Planta/crescimento & desenvolvimento , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/metabolismo
9.
Adv Healthc Mater ; 7(9): e1701163, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29283226

RESUMO

Synthetic cells, artificial cell-like particles, capable of autonomously synthesizing RNA and proteins based on a DNA template, are emerging platforms for studying cellular functions and for revealing the origins-of-life. Here, it is shown for the first time that artificial lipid-based vesicles, containing the molecular machinery necessary for transcription and translation, can be used to synthesize anticancer proteins inside tumors. The synthetic cells are engineered as stand-alone systems, sourcing nutrients from their biological microenvironment to trigger protein synthesis. When pre-loaded with template DNA, amino acids and energy-supplying molecules, up to 2 × 107 copies of green fluorescent protein are synthesized in each synthetic cell. A variety of proteins, having molecular weights reaching 66 kDa and with diagnostic and therapeutic activities, are synthesized inside the particles. Incubating synthetic cells, encoded to secrete Pseudomonas exotoxin A (PE) with 4T1 breast cancer cells in culture, resulted in killing of most of the malignant cells. In mice bearing 4T1 tumors, histological evaluation of the tumor tissue after a local injection of PE-producing particles indicates robust apoptosis. Synthetic cells are new platforms for synthesizing therapeutic proteins on-demand in diseased tissues.


Assuntos
ADP Ribose Transferases/biossíntese , Células Artificiais/metabolismo , Toxinas Bacterianas/biossíntese , Exotoxinas/biossíntese , Neoplasias Experimentais , Microambiente Tumoral , Fatores de Virulência/biossíntese , Animais , Linhagem Celular Tumoral , Feminino , Proteínas de Fluorescência Verde/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Exotoxina A de Pseudomonas aeruginosa
10.
ACS Nano ; 12(2): 1482-1490, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29365250

RESUMO

Surgical blades are common medical tools. However, blades cannot distinguish between healthy and diseased tissue, thereby creating unnecessary damage, lengthening recovery, and increasing pain. We propose that surgical procedures can rely on natural tissue remodeling tools-enzymes, which are the same tools our body uses to repair itself. Through a combination of nanotechnology and a controllably activated proteolytic enzyme, we performed a targeted surgical task in the oral cavity. More specifically, we engineered nanoparticles that contain collagenase in a deactivated form. Once placed at the surgical site, collagenase was released at a therapeutic concentration and activated by calcium, its biological cofactor that is naturally present in the tissue. Enhanced periodontal remodeling was recorded due to enzymatic cleavage of the supracrestal collagen fibers that connect the teeth to the underlying bone. When positioned in their new orientation, natural tissue repair mechanisms supported soft and hard tissue recovery and reduced tooth relapse. Through the combination of nanotechnology and proteolytic enzymes, localized surgical procedures can now be less invasive.


Assuntos
Colágeno/metabolismo , Colagenases/administração & dosagem , Colagenases/farmacologia , Tecido Conjuntivo/efeitos dos fármacos , Lipossomos/química , Nanopartículas/química , Animais , Colagenases/farmacocinética , Tecido Conjuntivo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Enzimas Imobilizadas/administração & dosagem , Enzimas Imobilizadas/farmacocinética , Enzimas Imobilizadas/farmacologia , Masculino , Boca/efeitos dos fármacos , Boca/metabolismo , Boca/cirurgia , Nanotecnologia/métodos , Proteólise/efeitos dos fármacos , Ratos Wistar
11.
J Control Release ; 257: 68-75, 2017 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-27744036

RESUMO

Injectable drug delivery systems that autonomously detect, propel towards, and ultimately treat the cancerous tissue, are the future of targeted medicine. Here, we developed a drug delivery system that swims autonomously towards cancer cells, where it releases a therapeutic cargo. This platform is based on viable bacteria, loaded with nanoparticles that contain the chemotherapeutic-antibiotic drug doxorubicin. The bacteria ferry across media and invade the cancer cells, increasing their velocity in the presence of nutrients that are present within the tumor microenvironment. Inside the cancer cells, doxorubicin is released from the nanoparticles, destroying the bacterial swimmer (antibiotic activity) and executing the therapeutic activity against the cancer cells (chemotherapeutic activity). This mode of delivery, where both the carrier and the cancer cell are destroyed, supports implementing nanoswimmers in drug delivery (Fig. 1).


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Escherichia coli/metabolismo , Neoplasias/tratamento farmacológico , Salmonella typhimurium/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Escherichia coli/citologia , Escherichia coli/efeitos dos fármacos , Lipossomos , Camundongos , Salmonella typhimurium/citologia , Salmonella typhimurium/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA