Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Proc Natl Acad Sci U S A ; 119(38): e2207525119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095208

ABSTRACT

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.


Subject(s)
Artificial Cells , Fibroblast Growth Factors , Neovascularization, Physiologic , Animals , Artificial Cells/transplantation , Cell Movement , Cell Proliferation , Collagen Type IV/metabolism , Endothelial Cells/physiology , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Humans , Mice , Protein Biosynthesis
2.
Chem Eng J ; 340: 9-14, 2018 May 15.
Article in English | MEDLINE | ID: mdl-31384170

ABSTRACT

Polylactic acid (PLA) is the most commonly used biodegradable polymer in clinical applications today. Examples range from drug delivery systems, tissue engineering, temporary and long-term implantable devices; constantly expanding to new fields. This is owed greatly to the polymer's favorable biocompatibility and to its safe degradation products. Once coming in contact with biological media, the polymer begins breaking down, usually by hydrolysis, into lactic acid (LA) or to carbon dioxide and water. These products are metabolized intracellularly or excreted in the urine and breath. Bacterial infection and foreign-body inflammation enhance the breakdown of PLA, through the secretion of enzymes that degrade the polymeric matrix. The biodegradation occurs both on the surface of the polymeric device and inside the polymer body, by diffusion of water between the polymer chains. The median half-life of the polymer is 30 weeks; however, this can be lengthened or shortened to address the clinical needs. Degradation kinetics can be tuned by determining the molecular composition and the physical architecture of the device. Using L- or D- chirality of the LA will greatly slow or lengthen the degradation rates, respectively. Despite the fact that this polymer is more than 150 years old, PLA remains a fertile platform for biomedical innovation and fundamental understanding of how artificial polymers can safely coexist with biological systems.

3.
Nanotechnology ; 28(43): 43LT01, 2017 Oct 27.
Article in English | MEDLINE | ID: mdl-28872058

ABSTRACT

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.


Subject(s)
Breast Neoplasms/diagnostic imaging , Drug Delivery Systems/methods , Liposomes/pharmacokinetics , Lung Neoplasms/diagnostic imaging , Neoplasm Metastasis/diagnostic imaging , Triple Negative Breast Neoplasms/diagnostic imaging , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/secondary , Cell Line, Tumor , Europium/chemistry , Europium/pharmacokinetics , Female , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Liposomes/chemical synthesis , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Optical Imaging , Rhodamines/chemistry , Rhodamines/pharmacokinetics , Tissue Distribution , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/secondary
4.
Adv Mater ; 35(51): e2304654, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37753928

ABSTRACT

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.


Subject(s)
Parkinson Disease , Animals , Humans , Mice , alpha-Synuclein/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Behavioral Symptoms , Brain/metabolism , Liposomes/metabolism , Parkinson Disease/drug therapy , Transferrins
5.
Nat Commun ; 13(1): 2328, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484097

ABSTRACT

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.


Subject(s)
Artificial Cells , Optogenetics , Light , Luciferases , Optogenetics/methods , Signal Transduction
6.
ACS Nano ; 16(4): 5246-5257, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35293714

ABSTRACT

Throughout the female menstrual cycle, physiological changes occur that affect the biodistribution of nanoparticles within the reproductive system. We demonstrate a 2-fold increase in nanoparticle accumulation in murine ovaries and uterus during ovulation, compared to the nonovulatory stage, following intravenous administration. This biodistribution pattern had positive or negative effects when drug-loaded nanoparticles, sized 100 nm or smaller, were used to treat different cancers. For example, treating ovarian cancer with nanomedicines during mouse ovulation resulted in higher drug accumulation in the ovaries, improving therapeutic efficacy. Conversely, treating breast cancer during ovulation, led to reduced therapeutic efficacy, due to enhanced nanoparticle accumulation in the reproductive system rather than at the tumor site. Moreover, chemotherapeutic nanoparticles administered during ovulation increased ovarian toxicity and decreased fertility compared to the free drug. The menstrual cycle should be accounted for when designing and implementing nanomedicines for females.


Subject(s)
Nanoparticles , Neoplasms , Female , Mice , Animals , Tissue Distribution , Fertility , Ovulation , Genitalia, Female
7.
Mhealth ; 7: 3, 2021.
Article in English | MEDLINE | ID: mdl-33634186

ABSTRACT

BACKGROUND: Interprofessional education (IPE) is a curricular requirement for all healthcare professional education standards. To foster learning about, from and with each other, consistent with the Interprofessional Education Consortium's Core Competencies, many graduate schools are integrating interprofessional (IP) simulation experiences throughout their educational curricula, providing multiple opportunities for health professional students to collaborate and practice together. High-fidelity, real-time simulations help students from diverse professional backgrounds to apply their classroom learning in realistic clinical situations, utilize mobile technology to access clinical decision support (CDS) software, and receive feedback in a safe setting, ensuring they are practice-ready upon graduation. METHODS: New York University Rory Meyers College of Nursing (NYU) and Long Island University College of Pharmacy (LIU) partnered for two consecutive years to create, coordinate and implement two interprofessional educational simulations involving patients with chronic cardiovascular disease. A utilization-focused evaluation of high-fidelity, simulation-enhanced IPE (Sim-IPE) was implemented to assess students' IP competencies before and after their participation in the IPE-simulation and their overall satisfaction with the experience. The Interprofessional Collaborative Competency Attainment Survey (ICCAS), a reliable instrument, was administered to both doctor of pharmacy students and primary care advanced practice nursing students before and after each simulation experience. Additionally, student satisfaction surveys were administered following the IPE-simulation. RESULTS: Aggregated means revealed statistically significant improvements in each of the six domains including communication, collaboration, roles and responsibilities, collaborative patient/family approach, conflict resolution and team functioning. Student ratings revealed positive experiences with the IPE-simulations. CONCLUSIONS: High-fidelity, real-time IPE-simulation is a powerful pedagogy to help graduate students from different professional backgrounds practice applying IP competencies in simulated experiences. Quality improvement studies and research studies are needed to assess the impact of high-fidelity, real-time simulations throughout graduate curricula with different types of patients to improve coordinated, team approaches to treatment.

8.
J Dent Educ ; 85(4): 504-512, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33230834

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the effectiveness of an annual oral-systemic health interprofessional education (IPE) clinical simulation and case study experience with nurse practitioner/midwifery (NP/MW), dental (DDS), medical (MD), and pharmacy (PharmD) students. METHODS: The Interprofessional Collaborative Competency Attainment Scale (ICCAS) was used to measure students' self-reported attainment of interprofessional competencies before and after the IPE experience. Pre- and post-test surveys were completed by NP/MW, DDS, MD, and PharmD student cohorts from 2017 to 2019. Students also had the opportunity to provide qualitative feedback about their experience at post-test. Data were collected from IPE faculty facilitators to assess their perception of the value of the Teaching Oral-Systemic Health (TOSH) program. RESULTS: Student ICCAS results demonstrated statistically significant improvement in self-reported interprofessional competencies among all types of students across all 3 years (P < 0.001); qualitative student comments reflected positive experiences with the TOSH program. Survey data from IPE faculty facilitators supported the value of the IPE experience for all students. CONCLUSIONS: The findings demonstrate the effectiveness of the TOSH program in using oral-systemic health as a clinical exemplar to develop interprofessional competencies. The 2017-2019 data reinforce the credibility of scaling the TOSH model for developing interprofessional competencies with students from different health professions.


Subject(s)
Nurse Practitioners , Oral Health , Curriculum , Education, Dental , Female , Humans , Interprofessional Relations , Pregnancy
9.
Sci Adv ; 7(41): eabj5435, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34613777

ABSTRACT

Neurons within the tumor microenvironment promote cancer progression; thus, their local targeting has potential clinical benefits. We designed PEGylated lipid nanoparticles loaded with a non-opioid analgesic, bupivacaine, to target neurons within breast cancer tumors and suppress nerve-to-cancer cross-talk. In vitro, 100-nm nanoparticles were taken up readily by primary neurons, trafficking from the neuronal body and along the axons. We demonstrate that signaling between triple-negative breast cancer cells (4T1) and neurons involves secretion of cytokines stimulating neurite outgrowth. Reciprocally, neurons stimulated 4T1 proliferation, migration, and survival through secretion of neurotransmitters. Bupivacaine curbs neurite growth and signaling with cancer cells, inhibiting cancer cell viability. In vivo, bupivacaine-loaded nanoparticles intravenously administered suppressed neurons in orthotopic triple-negative breast cancer tumors, inhibiting tumor growth and metastatic dissemination. Overall, our findings suggest that reducing nerve involvement in tumors is important for treating cancer.

10.
J Vis Exp ; (158)2020 04 27.
Article in English | MEDLINE | ID: mdl-32391815

ABSTRACT

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.


Subject(s)
Artificial Cells/metabolism , Emulsions/chemistry , Escherichia coli/metabolism , Protein Biosynthesis , Synthetic Biology/methods , Cell-Free System/metabolism , Green Fluorescent Proteins/metabolism , Liposomes/chemistry , Luciferases/metabolism
11.
Adv Mater ; 32(13): e1901989, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31286573

ABSTRACT

Artificial intelligence (AI) and nanotechnology are two fields that are instrumental in realizing the goal of precision medicine-tailoring the best treatment for each cancer patient. Recent conversion between these two fields is enabling better patient data acquisition and improved design of nanomaterials for precision cancer medicine. Diagnostic nanomaterials are used to assemble a patient-specific disease profile, which is then leveraged, through a set of therapeutic nanotechnologies, to improve the treatment outcome. However, high intratumor and interpatient heterogeneities make the rational design of diagnostic and therapeutic platforms, and analysis of their output, extremely difficult. Integration of AI approaches can bridge this gap, using pattern analysis and classification algorithms for improved diagnostic and therapeutic accuracy. Nanomedicine design also benefits from the application of AI, by optimizing material properties according to predicted interactions with the target drug, biological fluids, immune system, vasculature, and cell membranes, all affecting therapeutic efficacy. Here, fundamental concepts in AI are described and the contributions and promise of nanotechnology coupled with AI to the future of precision cancer medicine are reviewed.


Subject(s)
Artificial Intelligence , Nanomedicine/methods , Nanotechnology/methods , Precision Medicine/methods , Animals , Computational Biology/methods , Drug Delivery Systems/methods , Humans , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms/diagnosis , Neoplasms/therapy
12.
J Control Release ; 307: 331-341, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31238049

ABSTRACT

Lipid nanoparticles are used widely as anticancer drug and gene delivery systems. Internalizing into the target cell is a prerequisite for the proper activity of many nanoparticulate drugs. We show here, that the lipid composition of a nanoparticle affects its ability to internalize into triple-negative breast cancer cells. The lipid headgroup had the greatest effect on enhancing cellular uptake compared to other segments of the molecule. Having a receptor-targeted headgroup induced the greatest increase in cellular uptake, followed by cationic amine headgroups, both being superior to neutral (zwitterion) phosphatidylcholine or to negatively-charged headgroups. The lipid tails also affected the magnitude of cellular uptake. Longer acyl chains facilitated greater liposomal cellular uptake compared to shorter tails, 18:0 > 16:0 > 14:0. When having the same lipid tail length, unsaturated lipids were superior to saturated ones, 18:1 > 18:0. Interestingly, liposomes composed of phospholipids having 14:0 or 12:0-carbon-long-tails, such as DMPC and DLPC, decreased cell viability in a concertation dependent manner, due to a destabilizing effect these lipids had on the cancer cell membrane. Contrarily, liposomes composed of phospholipids having longer carbon tails (16:0 and 18:0), such as DPPC and HSPC, enhanced cancer cell proliferation. This effect is attributed to the integration of the exogenous liposomal lipids into the cancer-cell membrane, supporting the proliferation process. Cholesterol is a common lipid additive in nanoscale formulations, rigidifying the membrane and stabilizing its structure. Liposomes composed of DMPC (14:0) showed increased cellular uptake when enriched with cholesterol, both by endocytosis and by fusion. Contrarily, the effect of cholesterol on HSPC (18:0) liposomal uptake was minimal. Furthermore, the concentration of nanoparticles in solution affected their cellular uptake. The higher the concentration of nanoparticles the greater the absolute number of nanoparticles taken up per cell. However, the efficiency of nanoparticle uptake, i.e. the percent of nanoparticles taken up by cells, decreased as the concentration of nanoparticles increased. This study demonstrates that tuning the lipid composition and concentration of nanoscale drug delivery systems can be leveraged to modulate their cellular uptake.


Subject(s)
Drug Delivery Systems , Lipids/administration & dosage , Nanoparticles/administration & dosage , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocytosis , Lipids/chemistry , Mice , Nanoparticles/chemistry
13.
J Control Release ; 296: 1-13, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30615983

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Neoplasms , Sodium Bicarbonate/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Biological Transport/drug effects , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacokinetics , Female , Humans , Hydrogen-Ion Concentration , Liposomes , Mice, Inbred BALB C , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Sodium Bicarbonate/pharmacokinetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
ACS Nano ; 13(10): 11008-11021, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31503443

ABSTRACT

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.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Collagenases/pharmacology , Nanoparticles/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Collagen/chemistry , Collagen/genetics , Collagenases/chemistry , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Fibrosis/drug therapy , Fibrosis/pathology , Fibrosis/prevention & control , Humans , Liposomes/chemistry , Liposomes/pharmacology , Mice , Nanoparticles/therapeutic use , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pancreas/drug effects , Pancreas/pathology , Tumor Microenvironment/drug effects
15.
Sci Rep ; 8(1): 7589, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29773873

ABSTRACT

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.


Subject(s)
Crops, Agricultural/metabolism , Drug Delivery Systems , Liposomes/chemistry , Nanoparticles/administration & dosage , Nutrients/administration & dosage , Plant Leaves/metabolism , Solanum lycopersicum/metabolism , Crops, Agricultural/growth & development , Solanum lycopersicum/growth & development , Nanoparticles/chemistry , Plant Leaves/growth & development , Plant Roots/growth & development , Plant Roots/metabolism
16.
Adv Healthc Mater ; 7(9): e1701163, 2018 05.
Article in English | MEDLINE | ID: mdl-29283226

ABSTRACT

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.


Subject(s)
ADP Ribose Transferases/biosynthesis , Artificial Cells/metabolism , Bacterial Toxins/biosynthesis , Exotoxins/biosynthesis , Neoplasms, Experimental , Tumor Microenvironment , Virulence Factors/biosynthesis , Animals , Cell Line, Tumor , Female , Green Fluorescent Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Pseudomonas aeruginosa Exotoxin A
17.
ACS Nano ; 12(2): 1482-1490, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29365250

ABSTRACT

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.


Subject(s)
Collagen/metabolism , Collagenases/administration & dosage , Collagenases/pharmacology , Connective Tissue/drug effects , Liposomes/chemistry , Nanoparticles/chemistry , Animals , Collagenases/pharmacokinetics , Connective Tissue/metabolism , Drug Delivery Systems/methods , Enzymes, Immobilized/administration & dosage , Enzymes, Immobilized/pharmacokinetics , Enzymes, Immobilized/pharmacology , Male , Mouth/drug effects , Mouth/metabolism , Mouth/surgery , Nanotechnology/methods , Proteolysis/drug effects , Rats, Wistar
18.
J Control Release ; 257: 68-75, 2017 07 10.
Article in English | MEDLINE | ID: mdl-27744036

ABSTRACT

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).


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/metabolism , Drug Delivery Systems , Escherichia coli/metabolism , Neoplasms/drug therapy , Salmonella typhimurium/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Escherichia coli/cytology , Escherichia coli/drug effects , Liposomes , Mice , Salmonella typhimurium/cytology , Salmonella typhimurium/drug effects
19.
Nat Commun ; 7: 13325, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830705

ABSTRACT

Personalized medicine promises to revolutionize cancer therapy by matching the most effective treatment to the individual patient. Using a nanoparticle-based system, we predict the therapeutic potency of anticancer medicines in a personalized manner. We carry out the diagnostic stage through a multidrug screen performed inside the tumour, extracting drug activity information with single cell sensitivity. By using 100 nm liposomes, loaded with various cancer drugs and corresponding synthetic DNA barcodes, we find a correlation between the cell viability and the drug it was exposed to, according to the matching barcodes. Based on this screen, we devise a treatment protocol for mice bearing triple-negative breast-cancer tumours, and its results confirm the diagnostic prediction. We show that the use of nanotechnology in cancer care is effective for generating personalized treatment protocols.


Subject(s)
DNA/chemistry , Nanoparticles/chemistry , Precision Medicine/methods , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Base Sequence , Cell Line, Tumor , DNA/genetics , Drug Carriers/chemistry , Female , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
20.
PLoS One ; 11(10): e0165137, 2016.
Article in English | MEDLINE | ID: mdl-27768741

ABSTRACT

Cell-free protein synthesis (CFPS) systems are important laboratory tools that are used for various synthetic biology applications. Here, we present a simple and inexpensive laboratory-scale method for preparing a CFPS system from E. coli. The procedure uses basic lab equipment, a minimal set of reagents, and requires less than one hour to process the bacterial cell mass into a functional S30-T7 extract. BL21(DE3) and MRE600 E. coli strains were used to prepare the S30-T7 extract. The CFPS system was used to produce a set of fluorescent and therapeutic proteins of different molecular weights (up to 66 kDa). This system was able to produce 40-150 µg-protein/ml, with variations depending on the plasmid type, expressed protein and E. coli strain. Interestingly, the BL21-based CFPS exhibited stability and increased activity at 40 and 45°C. To the best of our knowledge, this is the most rapid and affordable lab-scale protocol for preparing a cell-free protein synthesis system, with high thermal stability and efficacy in producing therapeutic proteins.


Subject(s)
Escherichia coli Proteins/biosynthesis , Escherichia coli/metabolism , Cell-Free System
SELECTION OF CITATIONS
SEARCH DETAIL