Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cells ; 12(20)2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37887280

RESUMO

Mesothelial cells have been shown to have remarkable plasticity towards mesenchymal cell types during development and in disease situations. Here, we have characterized the potential of mesothelial cells to undergo changes toward perivascular cells using an in vitro angiogenesis assay. We demonstrate that GFP-labeled mesothelial cells (GFP-MCs) aligned closely and specifically with endothelial networks formed when human dermal microvascular endothelial cells (HDMECs) were cultured in the presence of VEGF-A165 on normal human dermal fibroblasts (NHDFs) for a 7-day period. The co-culture with GFP-MCs had a positive effect on branch point formation indicating that the cells supported endothelial tube formation. We interrogated the molecular response of the GFP-MCs to the angiogenic co-culture by qRT-PCR and found that the pericyte marker Ng2 was upregulated when the cells were co-cultured with HDMECs on NHDFs, indicating a change towards a perivascular phenotype. When GFP-MCs were cultured on the NHDF feeder layer, they upregulated the epithelial-mesenchymal transition marker Zeb1 and lost their circularity while increasing their size, indicating a change to a more migratory cell type. We analyzed the pericyte-like behavior of the GFP-MCs in a 3D cardiac microtissue (spheroid) with cardiomyocytes, cardiac fibroblasts and cardiac endothelial cells where the mesothelial cells showed alignment with the endothelial cells. These results indicate that mesothelial cells have the potential to adopt a perivascular phenotype and associate with endothelial cells to potentially support angiogenesis.


Assuntos
Células-Tronco Mesenquimais , Pericitos , Humanos , Células Endoteliais/metabolismo , Células Epiteliais , Técnicas de Cocultura
2.
Nanoscale Adv ; 5(20): 5520-5528, 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37822909

RESUMO

Tracking the biodistribution of cell therapies is crucial for understanding their safety and efficacy. Optical imaging techniques are particularly useful for tracking cells due to their clinical translatability and potential for intra-operative use to validate cell delivery. However, there is a lack of appropriate optical probes for cell tracking. The only FDA-approved material for clinical use is indocyanine green (ICG). ICG can be used for both fluorescence and photoacoustic imaging, but is prone to photodegradation, and at higher concentrations, undergoes quenching and can adversely affect cell health. We have developed novel near-infrared imaging probes comprising conjugated polymer nanoparticles (CPNs™) that can be fine-tuned to absorb and emit light at specific wavelengths. To compare the performance of the CPNs™ with ICG for in vivo cell tracking, labelled mesenchymal stromal cells (MSCs) were injected subcutaneously in mice and detected using fluorescence imaging (FI) and a form of photoacoustic imaging called multispectral optoacoustic tomography (MSOT). MSCs labelled with either ICG or CPN™ 770 could be detected with FI, but only CPN™ 770-labelled MSCs could be detected with MSOT. These results show that CPNs™ show great promise for tracking cells in vivo using optical imaging techniques, and for some applications, out-perform ICG.

3.
J Biophotonics ; 16(10): e202300109, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37431566

RESUMO

Tracking the fate of therapeutic cell types is important for assessing their safety and efficacy. Bioluminescence imaging (BLI) is an effective cell tracking technique, but poor spatial resolution means it has limited ability to precisely map cells in vivo in 3D. This can be overcome by using a bimodal imaging approach that combines BLI with a technique capable of generating high-resolution images. Here we compared the effectiveness of combining either multispectral optoacoustic tomography (MSOT) or micro-computed tomography (micro-CT) with BLI for tracking the fate of luciferase+ human mesenchymal stromal cells (MSCs) labelled with gold nanorods. Following subcutaneous administration in mice, the MSCs could be readily detected with MSOT but not with micro-CT. We conclude that MSOT is more sensitive than micro-CT for tracking gold nanorod-labelled cells in vivo and depending on the route of administration, can be used effectively with BLI to track MSC fate in mice.

4.
Stem Cell Res Ther ; 14(1): 120, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37143116

RESUMO

BACKGROUND: Mesenchymal stromal cells (MSCs), commonly sourced from adipose tissue, bone marrow and umbilical cord, have been widely used in many medical conditions due to their therapeutic potential. Yet, the still limited understanding of the underlying mechanisms of action hampers clinical translation. Clinical potency can vary considerably depending on tissue source, donor attributes, but importantly, also culture conditions. Lack of standard procedures hinders inter-study comparability and delays the progression of the field. The aim of this study was A- to assess the impact on MSC characteristics when different laboratories, performed analysis on the same MSC material using harmonised culture conditions and B- to understand source-specific differences. METHODS: Three independent institutions performed a head-to-head comparison of human-derived adipose (A-), bone marrow (BM-), and umbilical cord (UC-) MSCs using harmonised culture conditions. In each centre, cells from one specific tissue source were isolated and later distributed across the network to assess their biological properties, including cell expansion, immune phenotype, and tri-lineage differentiation (part A). To assess tissue-specific function, angiogenic and immunomodulatory properties and the in vivo biodistribution were compared in one expert lab (part B). RESULTS: By implementing a harmonised manufacturing workflow, we obtained largely reproducible results across three independent laboratories in part A of our study. Unique growth patterns and differentiation potential were observed for each tissue source, with similar trends observed between centres. Immune phenotyping verified expression of typical MSC surface markers and absence of contaminating surface markers. Depending on the established protocols in the different laboratories, quantitative data varied slightly. Functional experiments in part B concluded that conditioned media from BM-MSCs significantly enhanced tubulogenesis and endothelial migration in vitro. In contrast, immunomodulatory studies reported superior immunosuppressive abilities for A-MSCs. Biodistribution studies in healthy mice showed lung entrapment after administration of all three types of MSCs, with a significantly faster clearance of BM-MSCs. CONCLUSION: These results show the heterogeneous behaviour and regenerative properties of MSCs as a reflection of intrinsic tissue-origin properties while providing evidence that the use of harmonised culture procedures can reduce but do not eliminate inter-lab and operator differences.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Mesenquimais , Humanos , Animais , Camundongos , Células Cultivadas , Distribuição Tecidual , Diferenciação Celular , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células , Células da Medula Óssea , Cordão Umbilical
5.
Biomed Pharmacother ; 159: 114191, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36623449

RESUMO

Mesenchymal stromal cells (MSCs) are multipotent cells showing promise in pre-clinical studies and currently used in many clinical trials. The regenerative potential of MSCs is mediated, at least in part, by direct and indirect immunomodulatory processes. However, the mechanism of action is not fully understood yet, and there are still concerns about possible undesired negative effects associated with the administration of living cells. In this study, we (i) compare the long-term fate and safety of umbilical cord (UC-)MSCs administered to immunocompetent and immunocompromised (severe combined immunodeficient (SCID) and non-obese diabetic (NOD)/SCID) animals, and (ii) investigate the immunological response of the host to the administered cells. Intravenous administration of firefly luciferase expressing UC-MSCs revealed that the cells get trapped in the lungs of both immunocompetent and immunocompromised animals, with > 95% of the cells disappearing within 72 h after administration. In 27% of the SCID and 45% of the NOD/SCID, a small fraction of the cells lived up to day 14 but in most cases they all disappeared earlier. One NOD/SCID mouse showed a weak signal up to day 31. Immunocompetent mice displayed elevated percentages of neutrophils in the lungs, the blood, and the spleen 2 h after the administration of the cells. The concentration of neutrophil chemoattractants (MCP1, CCL7, Gro-α and IP-10) were also increased in the plasma of the animals 2 h after the administration of the MSCs. Our results suggest that although the UC-MSCs are short-lived in mice, they still result in an immunological response that might contribute to a therapeutic effect.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Cordão Umbilical , Sistema Imunitário , Células-Tronco Mesenquimais/fisiologia
6.
Mol Biotechnol ; 65(4): 544-555, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35999479

RESUMO

Genetic engineering of mesenchymal stromal cells (MSCs) is a tool widely used to explore MSC properties in vitro and in vivo. Lentiviral infection with the use of polycations as an adjuvant is a method that is commonly used to generate stably transduced cells. However, it is known that some polycations can negatively affect primary MSCs and to date, no study has explored the effect of different polycations on the transduction efficiency and properties of all main types of MSCs, namely those derived from umbilical cord, bone marrow and adipose tissue. Here we explore a range of polycations, using transduction protocols with and without spinoculation, to produce stably transduced MSCs from these three tissue sources. We identified that an overnight incubation with diethylaminoethyl-dextran (DEAE-Dextran) is the protocol associated with the best transduction efficiency without compromising the viability of the cells, and which worked consistently with lentiviral particles encoding for different transgenes. Transduced and sorted MSC populations revealed no significant changes in proliferation, morphology and expression of MSC markers compared to naïve MSCs. Following this study, we conclude that DEAE-Dextran is a polycation that can be successfully used to enhance the transduction of MSCs from all major tissue sources.


Assuntos
DEAE-Dextrano , Células-Tronco Mesenquimais , Humanos , Transdução Genética , DEAE-Dextrano/metabolismo , Lentivirus/genética , Vetores Genéticos/genética , Fenótipo , Proliferação de Células , Diferenciação Celular , Células Cultivadas
7.
Cancers (Basel) ; 14(5)2022 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-35267531

RESUMO

To investigate the utility of DCE-MRI derived pharmacokinetic parameters in evaluating tumour haemodynamic heterogeneity and treatment response in rodent models of glioblastoma, imaging was performed on intracranial F98 and GL261 glioblastoma bearing rodents. Clustering of the DCE-MRI-based parametric maps (using Tofts, extended Tofts, shutter speed, two-compartment, and the second generation shutter speed models) was performed using a hierarchical clustering algorithm, resulting in areas with poor fit (reflecting necrosis), low, medium, and high valued pixels representing parameters Ktrans, ve, Kep, vp, τi and Fp. There was a significant increase in the number of necrotic pixels with increasing tumour volume and a significant correlation between ve and tumour volume suggesting increased extracellular volume in larger tumours. In terms of therapeutic response in F98 rat GBMs, a sustained decrease in permeability and perfusion and a reduced cell density was observed during treatment with JAS239 based on Ktrans, Fp and ve as compared to control animals. No significant differences in these parameters were found for the GL261 tumour, indicating that this model may be less sensitive to JAS239 treatment regarding changes in vascular parameters. This study demonstrates that region-based clustered pharmacokinetic parameters derived from DCE-MRI may be useful in assessing tumour haemodynamic heterogeneity with the potential for assessing therapeutic response.

8.
Eur J Nucl Med Mol Imaging ; 49(3): 796-808, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34313817

RESUMO

INTRODUCTION: A novel, red-shifted bioluminescence imaging (BLI) system called AkaBLI has been recently developed for cell tracking in preclinical models and to date, limited data is available on how it performs in relation to existing systems. PURPOSE: To systematically compare the performance of AkaBLI and the standard Firefly luciferase (FLuc) systems to monitor the biodistribution and fate of cell therapies in rodents. METHODS: Umbilical cord mesenchymal stromal cells (MSCs) were transduced to produce two genetically engineered populations, expressing either AkaLuc or the engineered FLuc luc2. The bioluminescence of AkaLuc+ and FLuc+ cells was assessed both in vitro (emission spectra, saturation kinetics and light emission per cell) and in vivo (substrate kinetics following intraperitoneal and subcutaneous administration and biodistribution of the cells up to day 7). RESULTS: Introduction of the reporter genes has no effect on MSC phenotype. For BLI, the FLuc system is superior to AkaBLI in terms of (i) light output, producing a stronger signal after subcutaneous substrate delivery and more consistent signal kinetics when delivered intraperitoneally; (ii) absence of hepatic background; and (iii) safety, where the AkaLuc substrate was associated with a reaction in the skin of the mice in vivo. CONCLUSION: We conclude that there is no advantage in using the AkaBLI system to track the biodistribution of systemically administered cell-based regenerative medicine therapies in vivo.


Assuntos
Luciferases de Vaga-Lume , Células-Tronco Mesenquimais , Animais , Genes Reporter , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Distribuição Tecidual
9.
Emerg Top Life Sci ; 5(4): 549-562, 2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34495324

RESUMO

Mesenchymal stromal cells (MSCs) have been found to be safe and effective in a wide range of animal models of human disease. MSCs have been tested in thousands of clinical trials, but results show that while these cells appear to be safe, they tend to lack efficacy. This has raised questions about whether animal models are useful for predicting efficacy in patients. However, a problem with animal studies is that there is a lack of standardisation in the models and MSC therapy regimes used; there appears to be publication bias towards studies reporting positive outcomes; and the reproducibility of results from animal experiments tends not to be confirmed prior to clinical translation. A further problem is that while some progress has been made towards investigating the mechanisms of action (MoA) of MSCs, we still fail to understand how they work. To make progress, it is important to ensure that prior to clinical translation, the beneficial effects of MSCs in animal studies are real and can be repeated by independent research groups. We also need to understand the MoA of MSCs to assess whether their effects are likely to be beneficial across different species. In this review, we give an overview of the current clinical picture of MSC therapies and discuss what we have learned from animal studies. We also give a comprehensive update of what we know about the MoA of MSCs, particularly in relation to their role in immunomodulation.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Humanos , Imunidade , Imunomodulação , Transplante de Células-Tronco Mesenquimais/métodos , Reprodutibilidade dos Testes
10.
Magn Reson Med ; 86(1): 382-392, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33533114

RESUMO

PURPOSE: To establish high-frequency magnetic resonance electrical properties tomography (MREPT) as a novel contrast mechanism for the assessment of glioblastomas using a rat brain tumor model. METHODS: Six F98 intracranial tumor bearing rats were imaged longitudinally 8, 11 and 14 days after tumor cell inoculation. Conductivity and mean diffusivity maps were generated using MREPT and Diffusion Tensor Imaging. These maps were co-registered with T2 -weighted images and volumes of interests (VOIs) were segmented from the normal brain, ventricles, edema, viable tumor, tumor rim, and tumor core regions. Longitudinal changes in conductivity and mean diffusivity (MD) values were compared in these regions. A correlation analysis was also performed between conductivity and mean diffusivity values. RESULTS: The conductivity of ventricles, edematous area and tumor regions (tumor rim, viable tumor, tumor core) was significantly higher (P < .01) compared to the contralateral cortex. The conductivity of the tumor increased over time while MD from the tumor did not change. A marginal positive correlation was noted between conductivity and MD values for tumor rim and viable tumor, whereas this correlation was negative for the tumor core. CONCLUSION: We demonstrate a novel contrast mechanism based on ionic concentration and mobility, which may aid in providing complementary information to water diffusion in probing the microenvironment of brain tumors.


Assuntos
Neoplasias Encefálicas , Imagem de Tensor de Difusão , Animais , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/diagnóstico por imagem , Imagem de Difusão por Ressonância Magnética , Ratos , Tomografia , Microambiente Tumoral
11.
Dig Dis Sci ; 65(5): 1539-1548, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31571102

RESUMO

INTRODUCTION: Type 1 hepatorenal syndrome (HRS) is a fatal complication of cirrhosis. Treatments trend toward HRS reversal, but few show clear mortality benefit. We sought to quantify the progress-or lack thereof-in improving outcomes of type 1 HRS over time. METHODS: We performed a systematic review and meta-analysis for randomized controlled trials (RCTs) comparing type 1 HRS outcomes including (a) overall survival (liver transplant-free survival if reported) and (b) HRS reversal. Each study arm was analyzed separately to look at changes in outcomes over time. RCTs published comparing medical treatments for type 1 HRS were searched using several databases through July 31, 2019. RESULTS: Fourteen RCTs (28 arms) involving 778 participants enrolled between 2002 and 2018 were included. Twelve RCTs measured HRS reversal. In conjunction with albumin (or plasma expander), the most common medications used were terlipressin (13 arms), antibiotics (7), norepinephrine (6), dopamine (4), and midodrine/octreotide (3). Pooled survival rate was 34.6% (95% CI 26.4-43.8), and pooled HRS reversal rate was 42.8% (95% CI 34.2-51.9). Regression analyzing the incremental effect of the year the RCT was initiated showed that more recent studies were not associated with improved survival (OR 1.02, 95% CI 0.94-1.11, p = 0.66) or HRS reversal rates (OR 1.03, 95% CI 0.96-1.11, p = 0.41). There was no survival improvement when RCTs with endpoints assessed ≤ or > 1 month were analyzed separately with respective OR of 1.07 (95% CI 0.95-1.20, p = 0.26) and 0.97 (95% CI 0.85-1.12, p = 0.70). CONCLUSION: Outcomes have not improved for patients with type 1 HRS since 2002. There is a need to improve prevention and treatment of type 1 HRS.


Assuntos
Síndrome Hepatorrenal/tratamento farmacológico , Síndrome Hepatorrenal/mortalidade , Adulto , Albuminas/uso terapêutico , Antibacterianos/uso terapêutico , Dopamina/uso terapêutico , Quimioterapia Combinada , Feminino , Síndrome Hepatorrenal/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Midodrina/uso terapêutico , Norepinefrina/uso terapêutico , Octreotida/uso terapêutico , Substitutos do Plasma/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Análise de Regressão , Taxa de Sobrevida , Terlipressina/uso terapêutico , Resultado do Tratamento , Vasoconstritores/uso terapêutico , Adulto Jovem
12.
Mol Imaging Biol ; 22(4): 904-913, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31823201

RESUMO

PURPOSE: The question of whether mesenchymal stromal cells (MSCs) home to injured kidneys remains a contested issue. To try and understand the basis for contradictory findings reported in the literature, our purpose here was to investigate whether MSC homing capacity is influenced by administration route, the type of injury model used, and/or the presence of exogenous macrophages. PROCEDURES: To assess the viability, whole-body biodistribution, and intra-renal biodistribution of MSCs, we used a multimodal imaging strategy comprising bioluminescence and magnetic resonance imaging. The effect of administration route (venous or arterial) on the ability of MSCs to home to injured renal tissue, and persist there, was assessed in a glomerular injury model (induced by the nephrotoxicant, Adriamycin) and a tubular injury model induced by ischaemia-reperfusion injury (IRI). Exogenous macrophages were used as a positive control because these cells are known to home to injured mouse kidneys. To assess whether the homing capacity of MSCs can be influenced by the presence of exogenous macrophages, we used a dual-bioluminescence strategy that allowed the whole-body biodistribution of the two cell types to be monitored simultaneously in individual animals. RESULTS: Following intravenous administration, no MSCs were detected in the kidneys, irrespective of whether the mice had been subjected to renal injury. After arterial administration via the left cardiac ventricle, MSCs transiently populated the kidneys, but no preferential homing or persistence was observed in injured renal tissue after unilateral IRI. An exception was when MSCs were co-administered with exogenous macrophages; here, we observed some homing of MSCs to the injured kidney. CONCLUSIONS: Our findings strongly suggest that MSCs do not home to injured kidneys.


Assuntos
Rim/diagnóstico por imagem , Rim/lesões , Macrófagos/patologia , Células-Tronco Mesenquimais/patologia , Imagem Multimodal , Animais , Sobrevivência Celular , Rastreamento de Células , Modelos Animais de Doenças , Feminino , Fígado/diagnóstico por imagem , Medições Luminescentes , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Células RAW 264.7 , Traumatismo por Reperfusão/diagnóstico por imagem , Traumatismo por Reperfusão/patologia
13.
Nanoscale Adv ; 1(1): 367-377, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36132463

RESUMO

Nanoparticle contrast agents are useful tools to label stem cells and monitor the in vivo bio-distribution of labeled cells in pre-clinical models of disease. In this context, understanding the in vivo fate of the particles after injection of labelled cells is important for their eventual clinical use as well as for the interpretation of imaging results. We examined how the formulation of superparamagnetic iron oxide nanoparticles (SPIONs) impacts the labelling efficiency, magnetic characteristics and fate of the particles by comparing individual SPIONs with polyelectrolyte multilayer capsules containing SPIONs. At low labelling concentration, encapsulated SPIONs served as an efficient labelling agent for stem cells. The bio-distribution after intra-cardiac injection of labelled cells was monitored longitudinally by MRI and as an endpoint by inductively coupled plasma-optical emission spectrometry. The results suggest that, after being released from labelled cells after cell death, both formulations of particles are initially stored in liver and spleen and are not completely cleared from these organs 2 weeks post-injection.

14.
Stem Cell Res Ther ; 9(1): 332, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30486897

RESUMO

BACKGROUND: Cell-based regenerative medicine therapies are now frequently tested in clinical trials. In many conditions, cell therapies are administered systemically, but there is little understanding of their fate, and adverse events are often under-reported. Currently, it is only possible to assess safety and fate of cell therapies in preclinical studies, specifically by monitoring animals longitudinally using multi-modal imaging approaches. Here, using a suite of in vivo imaging modalities to explore the fate of a range of human and murine cells, we investigate how route of administration, cell type and host immune status affect the fate of administered cells. METHODS: We applied a unique imaging platform combining bioluminescence, optoacoustic and magnetic resonance imaging modalities to assess the safety of different human and murine cell types by following their biodistribution and persistence in mice following administration into the venous or arterial system. RESULTS: Longitudinal imaging analyses (i) suggested that the intra-arterial route may be more hazardous than intravenous administration for certain cell types, (ii) revealed that the potential of a mouse mesenchymal stem/stromal cell (MSC) line to form tumours depended on administration route and mouse strain and (iii) indicated that clinically tested human umbilical cord (hUC)-derived MSCs can transiently and unexpectedly proliferate when administered intravenously to mice. CONCLUSIONS: In order to perform an adequate safety assessment of potential cell-based therapies, a thorough understanding of cell biodistribution and fate post administration is required. The non-invasive imaging platform used here can expose not only the general organ distribution of these therapies, but also a detailed view of their presence within different organs and, importantly, tumourigenic potential. Our observation that the hUC-MSCs but not the human bone marrow (hBM)-derived MSCs persisted for a period in some animals suggests that therapies with these cells should proceed with caution.


Assuntos
Imageamento Tridimensional , Transplante de Células-Tronco Mesenquimais , Animais , Carcinogênese/patologia , Linhagem Celular , Humanos , Injeções Intravenosas , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos BALB C , Camundongos SCID , Osteossarcoma/patologia , Distribuição Tecidual , Cordão Umbilical/citologia
15.
Mol Imaging ; 17: 1536012118809585, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30392458

RESUMO

Metastasis is the most common cause of death for patients with cancer. To fully understand the steps involved in metastatic dissemination, in vivo models are required, of which murine ones are the most common. Therefore, preclinical imaging methods such as magnetic resonance imaging (MRI) have mainly been developed for small mammals and their potential to monitor cancer growth and metastasis in nonmammalian models is not fully harnessed. We have here used MRI to measure primary neuroblastoma tumor size and metastasis in a chick embryo model. We compared its sensitivity and accuracy to end-point fluorescence detection upon dissection. Human neuroblastoma cells labeled with green fluorescent protein (GFP) and micron-sized iron particles were implanted on the extraembryonic chorioallantoic membrane of the chick at E7. T2 RARE, T2-weighted fast low angle shot (FLASH) as well as time-of-flight MR angiography imaging were applied at E14. Micron-sized iron particle labeling of neuroblastoma cells allowed in ovo observation of the primary tumor and tumor volume measurement noninvasively. Moreover, T2 weighted and FLASH imaging permitted the detection of small metastatic deposits in the chick embryo, thereby reinforcing the potential of this convenient, 3R compliant, in vivo model for cancer research.


Assuntos
Imageamento por Ressonância Magnética , Metástase Neoplásica/diagnóstico por imagem , Metástase Neoplásica/patologia , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/patologia , Modelos Animais de Doenças , Desenvolvimento Embrionário , Humanos , Ferro/química , Metástase Neoplásica/diagnóstico , Tamanho da Partícula , Carga Tumoral
17.
Nanomedicine (Lond) ; 13(11): 1333-1348, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29949467

RESUMO

AIM: To develop a 3D neural cell construct for encapsulated delivery of transplant cells; develop hydrogels seeded with magnetic nanoparticle (MNP)-labeled cells suitable for cell tracking by MRI. MATERIALS & METHODS: Astrocytes were exogenously labeled with MRI-compatible iron-oxide MNPs prior to intra-construct incorporation within a 3D collagen hydrogel. RESULTS: A connective, complex cellular network was clearly observable within the 3D constructs, with high cellular viability. MNP accumulation in astrocytes provided a hypointense MRI signal at 24 h & 14 days. CONCLUSION: Our findings support the concept of developing a 3D construct possessing the dual advantages of (i) support of long-term cell survival of neural populations with (ii) the potential for noninvasive MRI-tracking of intra-construct cells for neuroregenerative applications.


Assuntos
Astrócitos/ultraestrutura , Terapia Baseada em Transplante de Células e Tecidos , Nanopartículas de Magnetita/administração & dosagem , Degeneração Neural/diagnóstico por imagem , Neurônios/ultraestrutura , Animais , Astrócitos/química , Astrócitos/transplante , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Rastreamento de Células , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/química , Microscopia Eletrônica de Transmissão , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Neurônios/química , Neurônios/transplante , Polímeros/administração & dosagem , Polímeros/química , Ratos
18.
Cancer Res ; 78(15): 4253-4269, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29789416

RESUMO

The ability of disseminated cancer cells to evade the immune response is a critical step for efficient metastatic progression. Protection against an immune attack is often provided by the tumor microenvironment that suppresses and excludes cytotoxic CD8+ T cells. Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive metastatic disease with unmet needs, yet the immunoprotective role of the metastatic tumor microenvironment in pancreatic cancer is not completely understood. In this study, we find that macrophage-derived granulin contributes to cytotoxic CD8+ T-cell exclusion in metastatic livers. Granulin expression by macrophages was induced in response to colony-stimulating factor 1. Genetic depletion of granulin reduced the formation of a fibrotic stroma, thereby allowing T-cell entry at the metastatic site. Although metastatic PDAC tumors are largely resistant to anti-PD-1 therapy, blockade of PD-1 in granulin-depleted tumors restored the antitumor immune defense and dramatically decreased metastatic tumor burden. These findings suggest that targeting granulin may serve as a potential therapeutic strategy to restore CD8+ T-cell infiltration in metastatic PDAC, thereby converting PDAC metastatic tumors, which are refractory to immune checkpoint inhibitors, into tumors that respond to immune checkpoint inhibition therapies.Significance: These findings uncover a mechanism by which metastatic PDAC tumors evade the immune response and provide the rationale for targeting granulin in combination with immune checkpoint inhibitors for the treatment of metastatic PDAC.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/15/4253/F1.large.jpg Cancer Res; 78(15); 4253-69. ©2018 AACR.


Assuntos
Resistencia a Medicamentos Antineoplásicos/fisiologia , Granulinas/metabolismo , Macrófagos/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Feminino , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/patologia , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral/fisiologia , Neoplasias Pancreáticas
19.
Contrast Media Mol Imaging ; 2018: 2514796, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30627058

RESUMO

The ability to track the biodistribution and fate of multiple cell populations administered to rodents has the potential to facilitate the understanding of biological processes in a range of fields including regenerative medicine, oncology, and host/pathogen interactions. Bioluminescence imaging is an important tool for achieving this goal, but current protocols rely on systems that have poor sensitivity or require spectral decomposition. Here, we show that a bioluminescence resonance energy transfer reporter (BRET) based on NanoLuc and LSSmOrange in combination with firefly luciferase enables the unambiguous discrimination of two cell populations in vivo with high sensitivity. We insert each of these reporter genes into cells using lentiviral vectors and demonstrate the ability to monitor the cells' biodistribution under a wide range of administration conditions, including the venous or arterial route, and in different tissues including the brain, liver, kidneys, and tumours. Our protocol allows for the imaging of two cell populations in the same imaging session, facilitating the overlay of the signals and the identification of anatomical positions where they colocalise. Finally, we provide a method for postmortem confirmation of the presence of each cell population in excised organs.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Medições Luminescentes/métodos , Imagem Óptica/métodos , Animais , Linhagem Celular , Genes Reporter , Vetores Genéticos , Luciferases de Vaga-Lume , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Distribuição Tecidual
20.
Biomater Sci ; 6(1): 101-106, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29188240

RESUMO

Although there is extensive literature covering the biomedical applications of superparamagnetic iron oxide nanoparticles (SPIONs), the phase of the iron oxide core used is not often taken into account when cell labelling and tracking studies for regenerative medicine are considered. Here, we use a co-precipitation reaction to synthesise particles of both magnetite- (Fe3O4) and maghemite- (γ-Fe2O3) based cores and consider whether the extra synthesis step to make maghemite based particles is advantageous for cell tracking.


Assuntos
Compostos Férricos/química , Nanopartículas de Magnetita/química , Meios de Contraste/química , Imageamento por Ressonância Magnética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...