Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Semin Cell Dev Biol ; 155(Pt C): 16-22, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-37479554

RESUMO

In the human body, the 1013 blood endothelial cells (ECs) which cover a surface of 500-700 m2 (Mai et al., 2013) are key players of tissue homeostasis, remodeling and regeneration. Blood vessel ECs play a major role in the regulation of metabolic and gaz exchanges, cell trafficking, blood coagulation, vascular tone, blood flow and fluid extravasation (also referred to as blood vascular permeability). ECs are heterogeneous in various capillary beds and have the exquisite capacity to cope with environmental changes by regulating their gene expression. Ischemia has major detrimental effects on the endothelium and ischemia-induced regulation of vascular integrity is of paramount importance for human health, as small amounts of fluid accumulation in the interstitium may be responsible for major effects on organ functions and patients outcome. In this review, we will here focus on the stimuli and the molecular mechanisms that control blood endothelium maintenance and phenotypic plasticity/transition involved in controlling blood capillary leakage that might open new avenues for therapeutic applications.


Assuntos
Células Endoteliais , Endotélio Vascular , Humanos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Isquemia/metabolismo , Permeabilidade Capilar , Adaptação Fisiológica , Permeabilidade
2.
Small ; 14(16): e1800020, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29542273

RESUMO

Once injected into a living organism, cells diffuse or migrate around the initial injection point and become impossible to be visualized and tracked in vivo. The present work concerns the development of a new technique for therapeutic cell labeling and subsequent in vivo visualization and magnetic retention. It is hypothesized and subsequently demonstrated that nanohybrids made of persistent luminescence nanoparticles and ultrasmall superparamagnetic iron oxide nanoparticles incorporated into a silica matrix can be used as an effective nanoplatform to label therapeutic cells in a nontoxic way in order to dynamically track them in real-time in vitro and in living mice. As a proof-of-concept, it is shown that once injected, these labeled cells can be visualized and attracted in vivo using a magnet. This first step suggests that these nanohybrids represent efficient multifunctional nanoprobes for further imaging guided cell therapies development.


Assuntos
Nanopartículas/química , Compostos Férricos/química , Luminescência
3.
Pharmacol Res ; 126: 123-137, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28720518

RESUMO

The structural complexity and physical properties of the tumor microenvironment negatively affect the penetration and efficiency of conventional anticancer drugs. While previously underestimated, the tumor microenvironment now becomes a potential target for cancer treatment. This microenvironment can be modulated either systemically by pharmacological means, or locally, through physical effects mediated by certain nanoparticles. Some of them, such as magnetic, plasmonic or carbon-based nanoparticles, can generate heat on demand in a spatially and temporally controlled manner. In addition, the nanoparticles can be either activated by light or magnetic stimuli. The impact of the resulting local heating can be observed on the ultrastructural level, as it strongly affects the organization of collagen fibers, and on the macroscopic level, since the thermal damages alter the mechanical properties of the tumor. Nanoparticle-based hyperthermia thus improves the effect of conventional anticancer drugs, as it allows their better penetration through the altered extracellular matrix. Here we suggest the use of nanoparticle-generated hyperthermia, obtained after magnetic or light activation, as an adjuvant treatment to prime the tumor microenvironment and improve the efficacy of chemotherapy.


Assuntos
Matriz Extracelular/efeitos dos fármacos , Febre/induzido quimicamente , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Humanos
4.
Nano Lett ; 12(9): 4830-7, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22928721

RESUMO

The fate of carbon nanotubes in the organism is still controversial. Here, we propose a statistical high-throughput imaging method to localize and quantify functionalized multiwalled carbon nanotubes in cells. We give the first experimental evidence of an intercellular translocation of carbon nanotubes. This stress-induced longitudinal transfer of nanomaterials is mediated by cell-released microvesicles known as vectors for intercellular communication. This finding raises new critical issues for nanotoxicology, since carbon nanotubes could be disseminated by circulating extracellular cell-released vesicles and visiting several cells in the course of their passage into the organism.


Assuntos
Citometria de Fluxo/métodos , Imagem Molecular/métodos , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestrutura , Frações Subcelulares/química , Frações Subcelulares/ultraestrutura , Células Cultivadas , Difusão , Humanos
5.
ACS Nano ; 15(2): 3251-3263, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33481565

RESUMO

The dissemination of tumor metastasis in the peritoneal cavity, also called peritoneal metastasis (PM) or carcinomatosis, represents a late stage of gastrointestinal and gynecological cancer with very poor prognosis, even when cytoreductive surgery is effective, due to residual microscopic disease. Photodynamic therapy (PDT) in the management of peritoneal metastasis has been clinically limited by the low tumor selectivity of photosensitizers (PS) and important adverse effects. Here, we propose extracellular nanovesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) as the fourth generation of immune active PS vectors that are able to target peritoneal metastasis with superior selectivity, potentiate PDT cytotoxicity at the tumor site without affecting healthy tissues, modulate the tumor microenvironment of immunocompetent colorectal and ovarian carcinomatosis models, and promote an antitumor immune response. A pioneering strategy was developed for high yield, large-scale production of MSC-EVs encapsulating the drug meta(tetrahydroxyphenyl)chlorin (mTHPC) (EVs-mTHPC) that is compatible with requirements of clinical translation and also preserves the topology and integrity of naturally produced EVs. Intraperitoneal injection of EVs-mTHPC showed an impressive enhancement of tumoral selectivity in comparison to the free drug and to the liposomal formulation Foslip (mean ratio of PS in tumors/organs of 40 for EVs-mTHPC versus 1.5 for the free PS and 5.5 for Foslip). PDT mediated by EVs-mTHPC permitted an important tumoral necrosis (55% of necrotic tumoral nodules versus 18% for Foslip (p < 0.0001)) and promoted antitumor immune cell infiltration, mainly proinflammatory M1-like CD80+ and CD8+ T cell effector. Intratumor proliferation was significantly decreased after PDT with EVs-mTHPC. Overall EVs vectorization of mTHPC afforded important tumoral selectivity while overcoming the PDT toxicity of the free drug and prolonged mice survival in the colorectal carcinomatosis model. MSC-EVs produced by our scalable manufacturing method appears like the clinically relevant fourth-generation PDT vehicle to overcome current limitations of PDT in the treatment of peritoneal metastasis and promote a hot tumor immune environment in PM.


Assuntos
Vesículas Extracelulares , Neoplasias Peritoneais , Fotoquimioterapia , Animais , Lipossomos , Mesoporfirinas , Camundongos , Neoplasias Peritoneais/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral
6.
Nanoscale ; 13(35): 14866-14878, 2021 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-34533159

RESUMO

In this study, we investigated the combination of extracellular (nano) vesicles (EVs) from pig adipose tissue-derived stromal cells (ADSCs) and a thermoresponsive gel, Pluronic® F-127 (PF-127), to prevent stricture formation after endoscopic resection in a porcine model. ADSC EVs were produced at a liter scale by a high-yielding turbulence approach from ADSCs 3D cultured in bioreactors and characterized in terms of size, morphology and membrane markers. The thermoresponsive property of the PF-127 gel was assessed by rheology. The pro-regenerative potency of ADSC EVs was investigated ex vivo in esophageal biopsies under starvation. In vivo tests were performed in a porcine model after extended esophageal endoscopic mucosal dissection (ESD). Pigs were randomized into 3 groups: control (n = 6), gel (n = 6) or a combination of 1.45 × 1012 EVs + gel (n = 6). Application of gel ± EVs was performed just after ESD with a follow-up finalized on day 21 post-ESD. There was a trend towards less feeding disorder in the EV + gel group in comparison with the gel and the control groups (16.67% vs. 66.7% vs. 83.33%, respectively) but without reaching a statistically significant difference. A significant decrease in the esophageal stricture rate was confirmed by endoscopic, radiological and histological examination for the EV + gel group. A decrease in the mean fibrosis area and larger regenerated muscularis mucosae were observed for the EV + gel group. In summary, the application of EVs + gel after extended esophageal endoscopic resection succeeded in preventing stricture formation with an anti-fibrotic effect. This nano-therapy may be of interest to tackle an unmet medical need considering that esophageal stricture is the most challenging delayed complication after extended superficial cancer resection by endoscopy.


Assuntos
Ressecção Endoscópica de Mucosa , Neoplasias Esofágicas , Estenose Esofágica , Vesículas Extracelulares , Animais , Tecido Adiposo , Hidrogéis/farmacologia , Células Estromais , Suínos
7.
Theranostics ; 7(2): 329-343, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28042338

RESUMO

Tumor stiffening, stemming from aberrant production and organization of extracellular matrix (ECM), has been considered a predictive marker of tumor malignancy, non-invasively assessed by ultrasound shear wave elastography (SWE). Being more than a passive marker, tumor stiffening restricts the delivery of diagnostic and therapeutic agents to the tumor and per se could modulate cellular mechano-signaling, tissue inflammation and tumor progression. Current strategies to modify the tumor extracellular matrix are based on ECM-targeting chemical agents but also showed deleterious systemic effects. On-demand excitable nanomaterials have shown their ability to perturb the tumor microenvironment in a spatiotemporal-controlled manner and synergistically with chemotherapy. Here, we investigated the evolution of tumor stiffness as well as tumor integrity and progression, under the effect of mild hyperthermia and thermal ablation generated by light-exposed multi-walled carbon nanotubes (MWCNTs) in an epidermoid carcinoma mouse xenograft. SWE was used for real-time mapping of the tumor stiffness, both during the two near infrared irradiation sessions and over the days after the treatment. We observed a transient and reversible stiffening of the tumor tissue during laser irradiation, which was lowered at the second session of mild hyperthermia or photoablation. In contrast, over the days following photothermal treatment, the treated tumors exhibited a significant softening together with volume reduction, whereas non-treated growing tumors showed an increase of tumor rigidity. The organization of the collagen matrix and the distribution of CNTs revealed a spatio-temporal correlation between the presence of nanoheaters and the damages on collagen and cells. This study highlights nanohyperthermia as a promising adjuvant strategy to reverse tumor stiffening and normalize the mechanical tumor environment.


Assuntos
Antineoplásicos/administração & dosagem , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Hipertermia Induzida/métodos , Nanoestruturas/administração & dosagem , Fotoquimioterapia/métodos , Animais , Modelos Animais de Doenças , Técnicas de Imagem por Elasticidade , Xenoenxertos , Lasers , Camundongos , Nanotubos de Carbono
8.
ACS Nano ; 9(8): 7925-39, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26168364

RESUMO

Safe implementation of nanotechnology and nanomedicine requires an in-depth understanding of the life cycle of nanoparticles in the body. Here, we investigate the long-term fate of gold/iron oxide heterostructures after intravenous injection in mice. We show these heterostructures degrade in vivo and that the magnetic and optical properties change during the degradation process. These particles eventually eliminate from the body. The comparison of two different coating shells for heterostructures, amphiphilic polymer or polyethylene glycol, reveals the long lasting impact of initial surface properties on the nanocrystal degradability and on the kinetics of elimination of magnetic iron and gold from liver and spleen. Modulation of nanoparticles reactivity to the biological environment by the choice of materials and surface functionalization may provide new directions in the design of multifunctional nanomedicines with predictable fate.


Assuntos
Envelhecimento/fisiologia , Materiais Revestidos Biocompatíveis/farmacocinética , Portadores de Fármacos/farmacocinética , Compostos Férricos/farmacocinética , Ouro/farmacocinética , Nanopartículas de Magnetita/análise , Alcenos/química , Animais , Materiais Revestidos Biocompatíveis/química , Portadores de Fármacos/química , Compostos Férricos/química , Ouro/química , Injeções Intravenosas , Fígado/metabolismo , Fígado/ultraestrutura , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestrutura , Anidridos Maleicos/química , Camundongos , Camundongos Endogâmicos C57BL , Nanomedicina/instrumentação , Nanomedicina/métodos , Polietilenoglicóis/química , Polímeros/química , Baço/metabolismo , Baço/ultraestrutura , Eletricidade Estática , Propriedades de Superfície
9.
ACS Nano ; 8(11): 11290-304, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25343751

RESUMO

Nanocomposites combining multiple functionalities in one single nano-object hold great promise for biomedical applications. In this work, carbon nanotubes (CNTs) were filled with ferrite nanoparticles (NPs) to develop the magnetic manipulation of the nanotubes and their theranostic applications. The challenges were both the filling of CNTs with a high amount of magnetic NPs and their functionalization to form biocompatible water suspensions. We propose here a filling process using CNTs as nanoreactors for high-yield in situ growth of ferrite NPs into the inner carbon cavity. At first, NPs were formed inside the nanotubes by thermal decomposition of an iron stearate precursor. A second filling step was then performed with iron or cobalt stearate precursors to enhance the encapsulation yield and block the formed NPs inside the tubes. Water suspensions were then obtained by addition of amino groups via the covalent functionalization of the external surface of the nanotubes. Microstructural and magnetic characterizations confirmed the confinement of NPs into the anisotropic structure of CNTs making them suitable for magnetic manipulations and MRI detection. Interactions of highly water-dispersible CNTs with tumor cells could be modulated by magnetic fields without toxicity, allowing control of their orientation within the cell and inducing submicron magnetic stirring. The magnetic properties were also used to quantify CNTs cellular uptake by measuring the cell magnetophoretic mobility. Finally, the photothermal ablation of tumor cells could be enhanced by magnetic stimulus, harnessing the hybrid properties of NP loaded-CNTs.


Assuntos
Diagnóstico por Imagem/instrumentação , Magnetismo , Nanopartículas Metálicas/química , Nanotubos de Carbono/química , Nanopartículas Metálicas/uso terapêutico , Microscopia Eletrônica de Transmissão , Óxidos/química
10.
ACS Nano ; 8(5): 4268-83, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24738788

RESUMO

Several studies propose nanoparticles for tumor treatment, yet little is known about the fate of nanoparticles and intimate interactions with the heterogeneous and ever-evolving tumor environment. The latter, rich in extracellular matrix, is responsible for poor penetration of therapeutics and represents a paramount issue in cancer therapy. Hence new strategies start aiming to modulate the neoplastic stroma. From this perspective, we assessed the efficacy of 19 nm PEG-coated iron oxide nanocubes with optimized magnetic properties to mediate mild tumor magnetic hyperthermia treatment. After injection of a low dose of nanocubes (700 µg of iron) into epidermoid carcinoma xenografts in mice, we monitored the effect of heating nanocubes on tumor environment. In comparison with the long-term fate after intravenous administration, we investigated spatiotemporal patterns of nanocube distribution, evaluated the evolution of cubes magnetic properties, and examined nanoparticle clearance and degradation processes. While inside tumors nanocubes retained their magnetic properties and heating capacity throughout the treatment due to a mainly interstitial extracellular location, the particles became inefficient heaters after cell internalization and transfer to spleen and liver. Our multiscale analysis reveals that collagen-rich tumor extracellular matrix confines the majority of nanocubes. However, nanocube-mediated hyperthermia has the potential to "destructure" this matrix and improve nanoparticle and drug penetration into neoplastic tissue. This study provides insight into dynamic interactions between nanoparticles and tumor components under physical stimulation and suggests that nanoparticle-mediated hyperthermia could be used to locally modify tumor stroma and thus improve drug penetration.


Assuntos
Compostos Férricos/química , Nanopartículas Metálicas/química , Neoplasias/patologia , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Colágeno/química , Espectroscopia de Ressonância de Spin Eletrônica , Matriz Extracelular/metabolismo , Feminino , Temperatura Alta , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Imageamento por Ressonância Magnética , Magnetismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Microscopia Eletrônica de Transmissão , Nanomedicina/métodos , Nanopartículas/química , Neoplasias/metabolismo , Polietilenoglicóis/química , Baço/efeitos dos fármacos , Baço/metabolismo
11.
J Vis Exp ; (82): e50566, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24378540

RESUMO

Carbon-based nanomaterials, like carbon nanotubes (CNTs), belong to this type of nanoparticles which are very difficult to discriminate from carbon-rich cell structures and de facto there is still no quantitative method to assess their distribution at cell and tissue levels. What we propose here is an innovative method allowing the detection and quantification of CNTs in cells using a multispectral imaging flow cytometer (ImageStream, Amnis). This newly developed device integrates both a high-throughput of cells and high resolution imaging, providing thus images for each cell directly in flow and therefore statistically relevant image analysis. Each cell image is acquired on bright-field (BF), dark-field (DF), and fluorescent channels, giving access respectively to the level and the distribution of light absorption, light scattered and fluorescence for each cell. The analysis consists then in a pixel-by-pixel comparison of each image, of the 7,000-10,000 cells acquired for each condition of the experiment. Localization and quantification of CNTs is made possible thanks to some particular intrinsic properties of CNTs: strong light absorbance and scattering; indeed CNTs appear as strongly absorbed dark spots on BF and bright spots on DF with a precise colocalization. This methodology could have a considerable impact on studies about interactions between nanomaterials and cells given that this protocol is applicable for a large range of nanomaterials, insofar as they are capable of absorbing (and/or scattering) strongly enough the light.


Assuntos
Citometria de Fluxo/métodos , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Nanotecnologia/métodos , Nanotubos de Carbono/análise , Células Cultivadas , Citometria de Fluxo/instrumentação , Fluoresceína-5-Isotiocianato/química , Corantes Fluorescentes/química , Humanos , Nanotecnologia/instrumentação
12.
ACS Nano ; 7(6): 4954-66, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23641799

RESUMO

Cell-released vesicles are natural carriers that circulate in body fluids and transport biological agents to distal cells. As nature uses vesicles in cell communication to promote tumor progression, we propose to harness their unique properties and exploit these biogenic carriers as Trojan horses to deliver therapeutic payloads to cancer cells. In a theranostic approach, cell-released vesicles were engineered by a top-down procedure from precursor cells, previously loaded with a photosensitizer and magnetic nanoparticles. The double exogenous cargo provided vesicles with magnetic and optical responsiveness allowing therapeutic and imaging functions. This new class of cell-derived smart nanovectors was named "theranosomes". Theranosomes enabled efficient photodynamic tumor therapy in a murine cancer model in vivo. Moreover the distribution of this biogenic vector could be monitored by dual-mode imaging, combining fluorescence and MRI. This study reports the first success in translating a cell communication mediator into a smart theranostic nanovector.


Assuntos
Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Imãs , Nanoestruturas , Neoplasias da Próstata/patologia , Neoplasias do Colo do Útero/patologia , Animais , Membrana Celular/metabolismo , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imageamento por Ressonância Magnética , Masculino , Mesoporfirinas/química , Mesoporfirinas/uso terapêutico , Camundongos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA