Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Res Sq ; 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38645111

RESUMO

Background: This study aimed to evaluate the effects of continuous administration of xylitol (a commonly used dental prebiotic) via a subcutaneous osmotic minipump in a B16F10 syngeneic mouse model. Methods: The B16F10 syngeneic model consisted of 6-8-week-old C57BL/6 male mice subcutaneously injected with five × 105 B16F10 cells suspended in 100 µl PBS in the right flank. The mice were randomly assigned to two groups: Group 1 was the treatment group with 10% xylitol-loaded pumps (n=10), while Group 2 was the control group with saline-loaded pumps (n=10). Alzet minipumps were implanted subcutaneously in the left flank of B16F10-injected mice once more than 50% of all mice developed palpable tumors. After pump implantation surgery, the mice were monitored daily and weighed 2-3x/week. Tumor sizes were measured with calipers 2-3x/week, and all mice were euthanized when their tumors became too large (20 mm on any axis or 2,000 mm3). The excised tumors were weighed and cut in half, with one half sent for histology and the other for metabolomic analysis. Results: The xylitol-treated group survived substantially longer than the control group. The tumor size was reduced by approximately 35% by volume. Histological sections of xylitol treat mice suggested reduced infiltration and angiogenesis, which is consistent with previous studies. The metabolomic analysis demonstrates that xylitol reduces the tumor production of histamine, NADP+, ATP, and glutathione from the tumor, thereby improving the host immune response with ROS reactive oxygen species. Conclusions: The results of this study suggest that xylitol has potential as an adjunct to oncological treatment and is being further investigated in comparison to monoclonal antibody therapy (Opdualag).

2.
Neuron ; 112(11): 1764-1777.e5, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38537641

RESUMO

Comprehensive, continuous quantitative monitoring of intricately orchestrated physiological processes and behavioral states in living organisms can yield essential data for elucidating the function of neural circuits under healthy and diseased conditions, for defining the effects of potential drugs and treatments, and for tracking disease progression and recovery. Here, we report a wireless, battery-free implantable device and a set of associated algorithms that enable continuous, multiparametric physio-behavioral monitoring in freely behaving small animals and interacting groups. Through advanced analytics approaches applied to mechano-acoustic signals of diverse body processes, the device yields heart rate, respiratory rate, physical activity, temperature, and behavioral states. Demonstrations in pharmacological, locomotor, and acute and social stress tests and in optogenetic studies offer unique insights into the coordination of physio-behavioral characteristics associated with healthy and perturbed states. This technology has broad utility in neuroscience, physiology, behavior, and other areas that rely on studies of freely moving, small animal models.


Assuntos
Comportamento Animal , Optogenética , Tecnologia sem Fio , Animais , Comportamento Animal/fisiologia , Optogenética/métodos , Camundongos , Frequência Cardíaca/fisiologia , Masculino , Próteses e Implantes , Taxa Respiratória/fisiologia , Monitorização Fisiológica/métodos , Monitorização Fisiológica/instrumentação , Algoritmos
3.
Nat Biomed Eng ; 7(10): 1252-1269, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37106153

RESUMO

Fully implantable wireless systems for the recording and modulation of neural circuits that do not require physical tethers or batteries allow for studies that demand the use of unconstrained and freely behaving animals in isolation or in social groups. Moreover, feedback-control algorithms that can be executed within such devices without the need for remote computing eliminate virtual tethers and any associated latencies. Here we report a wireless and battery-less technology of this type, implanted subdermally along the back of freely moving small animals, for the autonomous recording of electroencephalograms, electromyograms and body temperature, and for closed-loop neuromodulation via optogenetics and pharmacology. The device incorporates a system-on-a-chip with Bluetooth Low Energy for data transmission and a compressed deep-learning module for autonomous operation, that offers neurorecording capabilities matching those of gold-standard wired systems. We also show the use of the implant in studies of sleep-wake regulation and for the programmable closed-loop pharmacological suppression of epileptic seizures via feedback from electroencephalography. The technology can support a broader range of applications in neuroscience and in biomedical research with small animals.

4.
Proc Natl Acad Sci U S A ; 120(11): e2217734120, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36888661

RESUMO

Degradable polymer matrices and porous scaffolds provide powerful mechanisms for passive, sustained release of drugs relevant to the treatment of a broad range of diseases and conditions. Growing interest is in active control of pharmacokinetics tailored to the needs of the patient via programmable engineering platforms that include power sources, delivery mechanisms, communication hardware, and associated electronics, most typically in forms that require surgical extraction after a period of use. Here we report a light-controlled, self-powered technology that bypasses key disadvantages of these systems, in an overall design that is bioresorbable. Programmability relies on the use of an external light source to illuminate an implanted, wavelength-sensitive phototransistor to trigger a short circuit in an electrochemical cell structure that includes a metal gate valve as its anode. Consequent electrochemical corrosion eliminates the gate, thereby opening an underlying reservoir to release a dose of drugs by passive diffusion into surrounding tissue. A wavelength-division multiplexing strategy allows release to be programmed from any one or any arbitrary combination of a collection of reservoirs built into an integrated device. Studies of various bioresorbable electrode materials define the key considerations and guide optimized choices in designs. In vivo demonstrations of programmed release of lidocaine adjacent the sciatic nerves in rat models illustrate the functionality in the context of pain management, an essential aspect of patient care that could benefit from the results presented here.


Assuntos
Implantes Absorvíveis , Sistemas de Liberação de Medicamentos , Ratos , Animais , Eletrônica , Polímeros
5.
Science ; 376(6596): 1006-1012, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35617386

RESUMO

Temporary postoperative cardiac pacing requires devices with percutaneous leads and external wired power and control systems. This hardware introduces risks for infection, limitations on patient mobility, and requirements for surgical extraction procedures. Bioresorbable pacemakers mitigate some of these disadvantages, but they demand pairing with external, wired systems and secondary mechanisms for control. We present a transient closed-loop system that combines a time-synchronized, wireless network of skin-integrated devices with an advanced bioresorbable pacemaker to control cardiac rhythms, track cardiopulmonary status, provide multihaptic feedback, and enable transient operation with minimal patient burden. The result provides a range of autonomous, rate-adaptive cardiac pacing capabilities, as demonstrated in rat, canine, and human heart studies. This work establishes an engineering framework for closed-loop temporary electrotherapy using wirelessly linked, body-integrated bioelectronic devices.


Assuntos
Implantes Absorvíveis , Estimulação Cardíaca Artificial , Marca-Passo Artificial , Cuidados Pós-Operatórios , Tecnologia sem Fio , Animais , Cães , Frequência Cardíaca , Humanos , Cuidados Pós-Operatórios/instrumentação , Ratos
6.
Nat Commun ; 13(1): 3009, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35637230

RESUMO

Continuous, real-time monitoring of perfusion after microsurgical free tissue transfer or solid organ allotransplantation procedures can facilitate early diagnosis of and intervention for anastomotic thrombosis. Current technologies including Doppler systems, cutaneous O2-sensing probes, and fluorine magnetic resonance imaging methods are limited by their intermittent measurements, requirements for skilled personnel, indirect interfaces, and/or their tethered connections. This paper reports a wireless, miniaturized, minimally invasive near-infrared spectroscopic system designed for uninterrupted monitoring of local-tissue oxygenation. A bioresorbable barbed structure anchors the probe stably at implantation sites for a time period matched to the clinical need, with the ability for facile removal afterward. The probe connects to a skin-interfaced electronic module for wireless access to essential physiological parameters, including local tissue oxygenation, pulse oxygenation, and heart rate. In vitro tests and in vivo studies in porcine flap and kidney models demonstrate the ability of the system to continuously measure oxygenation with high accuracy and sensitivity.


Assuntos
Saturação de Oxigênio , Transplantes , Animais , Próteses e Implantes , Pele/diagnóstico por imagem , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Suínos
7.
Sci Adv ; 8(18): eabn4613, 2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35522741

RESUMO

Extensive antibody engineering and cloning is typically required to generate new bispecific antibodies. Made-to-order genes, advanced expression systems, and high-efficiency cloning can simplify and accelerate this process, but it still can take months before a functional product is realized. We developed a simple method to site-specifically and covalently attach a T cell-redirecting domain to any off-the-shelf, human immunoglobulin G (IgG) or native IgG isolated from serum. No antibody engineering, cloning, or knowledge of the antibody sequence is required. Bispecific antibodies are generated in just hours. By labeling antibodies isolated from tumor-bearing mice, including two syngeneic models, we generated T cell-redirecting autoantibodies (TRAAbs) that act as an effective therapeutic. TRAAbs preferentially bind tumor tissue over healthy tissue, indicating a previously unexplored therapeutic window. The use of autoantibodies to direct the tumor targeting of bispecific antibodies represents a new paradigm in personalized medicine that eliminates the need to identify tumor biomarkers.

8.
Nat Commun ; 11(1): 3273, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601301

RESUMO

Restoration of coronary blood flow after a heart attack can cause reperfusion injury potentially leading to impaired cardiac function, adverse tissue remodeling and heart failure. Iron is an essential biometal that may have a pathologic role in this process. There is a clinical need for a precise noninvasive method to detect iron for risk stratification of patients and therapy evaluation. Here, we report that magnetic susceptibility imaging in a large animal model shows an infarct paramagnetic shift associated with duration of coronary artery occlusion and the presence of iron. Iron validation techniques used include histology, immunohistochemistry, spectrometry and spectroscopy. Further mRNA analysis shows upregulation of ferritin and heme oxygenase. While conventional imaging corroborates the findings of iron deposition, magnetic susceptibility imaging has improved sensitivity to iron and mitigates confounding factors such as edema and fibrosis. Myocardial infarction patients receiving reperfusion therapy show magnetic susceptibility changes associated with hypokinetic myocardial wall motion and microvascular obstruction, demonstrating potential for clinical translation.


Assuntos
Ferro/análise , Traumatismo por Reperfusão Miocárdica/diagnóstico por imagem , Idoso , Animais , Estudos Transversais , Feminino , Ferritinas/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/patologia , Cicatrização
9.
J Am Chem Soc ; 142(17): 7783-7794, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32271558

RESUMO

Gold is a highly useful nanomaterial for many clinical applications, but its poor biodegradability can impair long-term physiological clearance. Large gold nanoparticles (∼10-200 nm), such as those required for long blood circulation times and appreciable tumor localization, often exhibit little to no dissolution and excretion. This can be improved by incorporating small gold particles within a larger entity, but elimination may still be protracted due to incomplete dispersion of gold. The present study describes a novel gold nanoparticle formulation capable of environmentally triggered decomposition. Ultrasmall gold nanoparticles are coated with thiolated dextran, and hydrophobic acetal groups are installed through direct covalent modification of the dextran. This hydrophobic exterior allows gold to be densely packed within ∼150 nm polymeric micelles. Upon exposure to an acidic environment, the acetal groups are cleaved and the gold nanoparticles become highly water-soluble, leading to destabilization of the micelle. Within 24 h, the ultrasmall water-soluble gold particles are released from the micelle and readily dispersed. Micelle degradation and gold nanoparticle dispersion was imaged in cultured macrophages, and micelle-treated mice displayed progressive physiological clearance of gold, with >85% elimination from the liver over three months. These particles present a novel nanomaterial formulation and address a critical unresolved barrier for clinical translation of gold nanoparticles.


Assuntos
Ouro/química , Interações Hidrofóbicas e Hidrofílicas , Nanopartículas Metálicas/química , Humanos , Concentração de Íons de Hidrogênio
10.
ACS Nano ; 14(1): 142-152, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31854966

RESUMO

Drug delivery to solid tumors is hindered by hydrostatic and physical barriers that limit the penetration of nanocarriers into tumor tissue. When exploiting the enhanced permeability and retention (EPR) effect for passive targeting of nanocarriers, the increased interstitial fluid pressure and dense extracellular matrix in tumors limits the distribution of the nanocarriers to perivascular regions. Previous strategies have shown that magnetophoresis enhances accumulation and penetration of nanoparticles into solid tumors. However, because magnetic fields fall off rapidly with distance from the magnet, these methods have been limited to use in superficial tumors. To overcome this problem, we have developed a system comprising two oppositely polarized magnets that enables the penetration of magnetic nanocarriers into more deeply seeded tumors. Using this method, we demonstrate a 5-fold increase in the penetration and a 3-fold increase in the accumulation of magnetic nanoparticles within solid tumors compared to EPR.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Nanopartículas/química , Animais , Neoplasias da Mama/patologia , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Feminino , Fenômenos Magnéticos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/patologia , Tamanho da Partícula , Propriedades de Superfície
11.
Adv Ther (Weinh) ; 2(9)2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35494480

RESUMO

Traditional oncology treatment modalities are often associated with a poor therapeutic index. This has driven the development of new targeted treatment modalities, including several based on the conversion of optical light into heat energy (photothermal therapy, PTT) and sound waves (photoacoustic imaging, PA) that can be applied locally. These approaches are especially effective when combined with photoactive nanoparticles that preferentially accumulate in tissues of interest and thereby further increase spatiotemporal resolution. In this study, two clinically-used materials that have proven effective in both PTT and PA - indocyanine green and gold nanoparticles - were combined into a single nanoformulation. These particles, "ICG-AuNP clusters", incorporated high concentrations of both moieties without the need for additional stabilizing or solubilizing reagents. The clusters demonstrated high theranostic efficacy both in vitro and in vivo, compared with ICG alone. Specifically, in an orthotopic mouse model of triple-negative breast cancer, ICG-AuNP clusters could be injected intravenously, imaged in the tumor by PA, and then combined with near-infrared laser irradiation to successfully thermally ablate tumors and prolong animal survival. Altogether, this novel nanomaterial demonstrates excellent therapeutic potential for integrated treatment and imaging.

12.
J Assoc Res Otolaryngol ; 19(2): 123-132, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29349595

RESUMO

Cisplatin-induced hearing loss is experienced by a high percentage of patients with squamous cell carcinoma undergoing cisplatin chemotherapy. A novel nano-construct capable of sequestering extracellular cisplatin was developed to combat this problem. The nano-construct consisted of superparamagnetic iron oxide nanoparticles (SPIONs) entrapped within polymeric micelles, which were formed from a glutathione diethyl ester-conjugated amphiphilic diblock copolymer. The glutathione-micelles were analyzed at the cellular level and in an organotypic study for safety evaluation. All utilized methods indicated that the micelles do not cause cellular toxicity or organ damage. The micelles' ability to reduce cisplatin-induced cytotoxicity was then probed in an in vitro model. Cisplatin was pre-treated with the novel nano-construct before being added to growing cells. When compared to cells that were exposed to untreated cisplatin, cells in the pre-treated cisplatin group showed a significant increase in cell viability. This clearly demonstrates that the construct is able to protect the cells from cisplatin cytotoxicity and makes it highly likely that the novel nano-construct will be able to play a role in the protection of the inner ear from cisplatin-induced ototoxicity.


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Glutationa/uso terapêutico , Perda Auditiva/prevenção & controle , Nanopartículas Metálicas/uso terapêutico , Animais , Antineoplásicos/química , Cisplatino/química , Avaliação Pré-Clínica de Medicamentos , Glutationa/química , Nanopartículas Metálicas/química , Camundongos , Micelas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...