Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Transl Stroke Res ; 14(4): 545-561, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35900719

RESUMO

Compromised adult human mesenchymal stem cells (hMSC) can impair cell therapy efficacy and further reverse ischemic recovery. However, in vitro assays require extended passage to characterize cells, limiting rapid assessment for therapeutic potency. Multinuclear magnetic resonance imaging and spectroscopy (MRI/S) provides near real-time feedback on disease progression and tissue recovery. Applied to ischemic stroke, 23Na MRI evaluates treatment efficacy within 24 h after middle cerebral artery occlusion, showing recovery of sodium homeostasis and lesion reduction in specimens treated with hMSC while 1H MRS identifies reduction in lactate levels. This combined metric was confirmed by evaluating treatment groups receiving healthy or compromised hMSC versus vehicle (sham saline injection) over 21 days. Behavioral tests to assess functional recovery and cell analysis for immunomodulatory and macrophage activity to detect hMSC potency confirm MR findings. Clinically, these MR metrics may prove critical to early evaluations of therapeutic efficacy and overall stroke recovery.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Adulto , Humanos , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/terapia , Acidente Vascular Cerebral/patologia , Infarto da Artéria Cerebral Média/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Imageamento por Ressonância Magnética/métodos
2.
Biomedicines ; 10(12)2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36551816

RESUMO

The significant roles of extracellular vesicles (EVs) as intracellular mediators, disease biomarkers, and therapeutic agents, make them a scientific hotspot. In particular, EVs secreted by human stem cells show significance in treating neurological disorders, such as Alzheimer's disease and ischemic stroke. However, the clinical applications of EVs are limited due to their poor targeting capabilities and low therapeutic efficacies after intravenous administration. Superparamagnetic iron oxide (SPIO) nanoparticles are biocompatible and have been shown to improve the targeting ability of EVs. In particular, ultrasmall SPIO (USPIO, <50 nm) are more suitable for labeling nanoscale EVs due to their small size. In this study, induced forebrain neural progenitor cortical organoids (iNPCo) were differentiated from human induced pluripotent stem cells (iPSCs), and the iNPCo expressed FOXG1, Nkx2.1, α-catenin, as well as ß-tubulin III. EVs were isolated from iNPCo media, then loaded with USPIOs by sonication. Size and concentration of EV particles were measured by nanoparticle tracking analysis, and no significant changes were observed in size distribution before and after sonication, but the concentration decreased after labeling. miR-21 and miR-133b decreased after sonication. Magnetic resonance imaging (MRI) demonstrated contrast visualized for the USPIO labeled EVs embedded in agarose gel phantoms. Upon calculation, USPIO labeled EVs exhibited considerably shorter relaxation times, quantified as T2 and T2* values, reducing the signal intensity and generating higher MRI contrast compared to unlabeled EVs and gel only. Our study demonstrated that USPIO labeling was a feasible approach for in vitro tracking of brain organoid-derived EVs, which paves the way for further in vivo examination.

3.
J Extracell Vesicles ; 11(6): e12235, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35716062

RESUMO

Human mesenchymal stem cell (hMSC) derived extracellular vesicles (EVs) have shown therapeutic potential in recent studies. However, the corresponding therapeutic components are largely unknown, and scale-up production of hMSC EVs is a major challenge for translational applications. In the current study, hMSCs were grown as 3D aggregates under wave motion to promote EV secretion. Results demonstrate that 3D hMSC aggregates promote activation of the endosomal sorting complexes required for transport (ESCRT)-dependent and -independent pathways. mRNA sequencing revealed global transcriptome alterations for 3D hMSC aggregates. Compared to 2D-hMSC-EVs, the quantity of 3D-hMSC-EVs was enhanced significantly (by 2-fold), with smaller sizes, higher miR-21 and miR-22 expression, and an altered protein cargo (e.g., upregulation of cytokines and anti-inflammatory factors) uncovered by proteomics analysis, possibly due to altered EV biogenesis. Functionally, 3D-hMSC-EVs rejuvenated senescent stem cells and exhibited enhanced immunomodulatory potentials. In summary, this study provides a promising strategy for scalable production of high-quality EVs from hMSCs with enhanced therapeutic potential.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Comunicação Celular , Vesículas Extracelulares/metabolismo , Humanos , MicroRNAs/metabolismo , Proteômica/métodos
4.
Transl Stroke Res ; 13(4): 543-555, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35132543

RESUMO

Extended therapeutic application remains a significant issue in the use of stem cell therapies to treat ischemic stroke. Along these lines, neurological recovery in a rodent model of ischemic stroke was evaluated following implantation of human mesenchymal stem cell aggregates (hMSC-agg), labeled with micron-sized particles of iron oxide, directly into the lateral ventricle contralateral to the ischemic lesion hemisphere. Longitudinally, disease progression and response to hMSC-agg therapy were assessed by 1H and 23Na magnetic resonance imaging (MRI) at 21.1 T to investigate cellular localization, migration, and recovery over an extended timeframe. MRI provides quantifiable metrics of tissue status through sodium distributions in addition to traditional proton imaging. Quantitative 23Na MRI revealed a significant decrease of sodium concentrations following hMSC aggregate implantation, indicating recovery of homeostasis. This result correlates positively with extended neurological recovery assessed by behavioral analysis and immunohistochemistry. These findings demonstrate the potential of implanted hMSC aggregate therapy to provide extended treatment for ischemic stroke, as well as the robustness of MRI for monitoring such approaches. This method potentially can be translated to a clinical setting for the assessment of extended cell therapy efficacy.


Assuntos
AVC Isquêmico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Isquemia/metabolismo , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Sódio/metabolismo , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/cirurgia
5.
Magn Reson Med ; 86(6): 3211-3223, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34355818

RESUMO

PURPOSE: Diffusion MRI offers insight into ischemic stroke progression in both human and rodent models. However, diffusion MRI to evaluate therapeutic application of mesenchymal stem cells is limited. Robust analytical techniques are required to identify potential physiological changes as a function of cell therapy in stroke. Here, we seek to establish Neurite Orientation Dispersion and Density Imaging (NODDI) as a feasible method in evaluating stroke evolution in response to cell-based therapeutics. METHODS: Diffusion MRI data at 21.1T were acquired from 16 male rats. Rats were grouped randomly: naïve (baseline, N = 5), stroke with injections of phosphate buffered saline (N = 6), stroke with injection of 2D human mesenchymal stem cells (hMSC, N = 5). Data were acquired on days 1, 3, 7, and 21 post-surgery. DTI and NODDI maps were generated, with regions of interest placed in the ischemic hemisphere external capsule and striatum. Diffusion parameters were compared between groups each day, and within groups across hemispheres and longitudinally. Behavioral characterizations were on days 0 (pre-surgery), 3, 7, 14, and 21. RESULTS: The 2D hMSC preserved diffusional restriction in the external capsule compared to saline (day 1: MD, P = .4060; AD, P = .0220). NODDI indicates that hMSC may have preserved intracellular volume fractions (ICVF: day 1, P = .0086; day 3, P = .0021; day 21, P = .0383). Diffusion metrics of hMSC treated animals were comparable to naïve for the external capsule. CONCLUSIONS: NODDI compliments DTI metrics, enhances interpretation of tissue outcome in ischemic stroke following hMSC application, and may be useful in evaluating or predicting therapeutic response.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Substância Branca , Animais , Encéfalo , Isquemia Encefálica/diagnóstico por imagem , Imagem de Difusão por Ressonância Magnética , Imagem de Tensor de Difusão , Substância Cinzenta , Humanos , Masculino , Neuritos , Ratos , Células-Tronco , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/cirurgia
6.
Theranostics ; 11(17): 8129-8142, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373732

RESUMO

Alzheimer's disease (AD) is an irreversible neurodegenerative disorder that affects more than 44 million people worldwide. Despite the high disease burden, there is no effective treatment for people suffering from AD. Mesenchymal stem cells (MSCs) are multipotent stromal cells that have been widely studied due to their therapeutic potential. However, administration of cells has been found to have a multitude of limitations. Recently, extracellular vesicles (EVs) derived from MSCs have been studied as a therapeutic candidate, as they exhibit similar immunoprotective and immunomodulatory abilities as the host human MSCs. Methods: To test the potential therapeutic effects of MSC EVs, human bone-marrow derived MSCs were grown in three-dimensional (3D) cell culture, and small EVs were harvested using differential ultracentrifugation. These small EVs were given to non-transgenic (NT) or 5XFAD (5 familial Alzheimer's disease mutations) mice intranasally (IN) every 4 days for 4 months. The mice were then required to perform a variety of behavioral assays to measure changes in learning and memory. Afterwards, immunohistochemistry was performed on brain slices to measure amyloid beta (Aß) and glial fibrillary acidic protein (GFAP) levels. Results: The data revealed that 5XFAD mice that received hMSC-EV treatment behaved significantly better in cognitive tests than saline treated 5XFAD mice, with no significant change between EV-treated 5XFAD mice and NT mice. Additionally, we found lower Aß plaque load in the hippocampus of the EV-treated mice. Finally, less colocalization between GFAP and Aß plaques was found in the brain of EV-treated mice compared to saline. Conclusions: Taken together, these data suggest that IN administration of MSC-derived EVs can slow down AD pathogenesis.


Assuntos
Doença de Alzheimer/terapia , Transplante de Células-Tronco Mesenquimais , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Hipocampo/metabolismo , Imunomodulação , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo
7.
ACS Biomater Sci Eng ; 7(3): 1111-1122, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33525864

RESUMO

Stem-cell-derived extracellular vesicles (EVs) are promising tools for therapeutic delivery and imaging in the medical research fields. EVs that arise from endosomal compartments or plasma membrane budding consist of exosomes and microvesicles, which range between 30 and 200 nm and 100-1000 nm, respectively. Iron oxide nanoparticles can be used to label stem cells or possibly EVs for magnetic resonance imaging. This could be a novel way to visualize areas in the body that are affected by neurological disorders such as stroke. Human induced pluripotent stem cells (iPSK3 cells) were plated on low-attachment plates and treated with SB431542 and LDN193189 during the first week for the induction of cortical spheroid formation and grown with fibroblast growth factor 2 and cyclopamine during the second week for the neural progenitor cell (iNPC) differentiation. iNPCs were then grown on attachment plates and treated with iron oxide (Fe3O4) nanoparticles at different sizes (8, 15, and 30 nm in diameter) and concentrations (0.1, 10, and 100 µM). The spheroids and media collected from these cultures were used for iron oxide detection as well as EV isolation and characterizations, respectively. MTT assay demonstrated that the increased size and concentration of the iron oxide nanoparticles had little effect on the metabolic activity of iNPCs. In addition, the Live/Dead assay showed high viability in all the nanoparticle treated groups and the untreated control. The EVs isolated from these culture groups were analyzed and displayed similar or higher EV counts compared with control. The observed EV size averaged 200-250 nm, and electron microscopy revealed the expected exosome morphology for EVs from all groups. RT-PCR analysis of EV biogenesis markers (CD63, CD81, Alix, TSG101, Syntenin1, ADAM10, RAB27b, and Syndecan) showed differential expression between the iron-oxide-treated cultures and nontreated cultures, as well as between adherent and nonadherent 3D cultures. Iron oxide nanoparticles were detected inside the cortical spheroid cells but not EVs by MRI. The addition of iron oxide nanoparticles does not induce significant cytotoxic effects to cortical spheroids. In addition,, nanoparticles may stimulate the biogenesis of EVs when added to cortical spheroids in vitro.


Assuntos
Vesículas Extracelulares , Células-Tronco Pluripotentes Induzidas , Compostos Férricos , Humanos , Ferro , Óxidos
8.
Sci Rep ; 10(1): 20448, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33235227

RESUMO

Mesenchymal stem cell (MSC)-based therapy has shown great promises in various animal disease models. However, this therapeutic potency has not been well claimed when applied to human clinical trials. This is due to both the availability of MSCs at the time of administration and lack of viable expansion strategies. MSCs are very susceptible to in vitro culture environment and tend to adapt the microenvironment which could lead to cellular senescence and aging. Therefore, extended in vitro expansion induces loss of MSC functionality and its clinical relevance. To combat this effect, this work assessed a novel cyclical aggregation as a means of expanding MSCs to maintain stem cell functionality. The cyclical aggregation consists of an aggregation phase and an expansion phase by replating the dissociated MSC aggregates onto planar tissue culture surfaces. The results indicate that cyclical aggregation maintains proliferative capability, stem cell proteins, and clonogenicity, and prevents the acquisition of senescence. To determine why aggregation was responsible for this phenomenon, the integrated stress response pathway was probed with salubrial and GSK-2606414. Treatment with salubrial had no significant effect, while GSK-2606414 mitigated the effects of aggregation leading to in vitro aging. This method holds the potential to increase the clinical relevance of MSC therapeutic effects from small model systems (such as rats and mice) to humans, and may open the potential of patient-derived MSCs for treatment thereby removing the need for immunosuppression.


Assuntos
Adenina/análogos & derivados , Técnicas de Cultura de Células/métodos , Cinamatos/farmacologia , Indóis/farmacologia , Células-Tronco Mesenquimais/citologia , Tioureia/análogos & derivados , Adenina/farmacologia , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Humanos , Propriedades de Superfície , Tioureia/farmacologia
9.
Biotechnol Bioeng ; 117(10): 3136-3149, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32579299

RESUMO

Protein homeostasis is critical for cellular function, as loss of homeostasis is attributed to aging and the accumulation of unwanted proteins. Human mesenchymal stem cells (MSCs) have shown promising therapeutic potential due to their impressive abilities to secrete inflammatory modulators, angiogenic, and regenerative cytokines. However, there exists the problem of human MSC expansion with compromised therapeutic quality. Duringin vitro expansion, human MSCs are plated on stiff plastics and undergo culture adaptation, which results in aberrant proliferation, shifts in metabolism, and decreased autophagic activity. It has previously been shown that three-dimensional (3D) aggregation can reverse some of these alterations by heightening autophagy and recovering the metabolic state back to a naïve phenotype. To further understand the proteostasis in human MSC culture, this study investigated the effects of 3D aggregation on the human MSC proteome to determine the specific pathways altered by aggregation. The 3D aggregates and 2D cultures of human MSCs derived from bone marrow (bMSC) and adipose tissue (ASC) were analyzed along with differentiated human dermal fibroblasts (FB). The proteomics analysis showed the elevated eukaryotic initiation factor 2 pathway and the upregulated activity of the integrated stress response (ISR) in 3D aggregates. Specific protein quantification further determined that bMSC and ASC responded to ISR, while FB did not. 3D aggregation significantly increased the ischemic survival of bMSCs and ASCs. Perturbation of ISR with small molecules salubrinal and GSK2606414 resulted in differential responses of bMSC, ASC, and FB. This study indicates that aggregation-based preconditioning culture holds the potential for improving the therapeutic efficacy of expanded human MSCs via the establishment of ISR and homeostasis.


Assuntos
Tecido Adiposo/citologia , Medula Óssea/metabolismo , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Agregação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Estresse Fisiológico
10.
NMR Biomed ; 33(2): e4186, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31797472

RESUMO

MRI leverages multiple modes of contrast to characterize stroke. High-magnetic-field systems enhance the performance of these MRI measurements. Previously, we have demonstrated that individually sodium and stem cell tracking metrics are enhanced at ultrahigh field in a rat model of stroke, and we have developed robust single-scan diffusion-weighted imaging approaches that utilize spatiotemporal encoding (SPEN) of the apparent diffusion coefficient (ADC) for these challenging field strengths. Here, we performed a multiparametric study of middle cerebral artery occlusion (MCAO) biomarker evolution focusing on comparison of these MRI biomarkers for stroke assessment during sub-acute recovery in rat MCAO models at 21.1 T. T2 -weighted MRI was used as the benchmark for identification of the ischemic lesion over the course of the study. The number of MPIO-induced voids measured by gradient-recalled echo, the SPEN measurement of ADC, and 23 Na MRI values were determined in the ischemic area and contralateral hemisphere, and relative performances for stroke classification were compared by receiver operator characteristic analysis. These measurements were associated with unique time-dependent trajectories during stroke recovery that changed the sensitivity and specificity for stroke monitoring during its evolution. Advantages and limitations of these contrasts, and the use of ultrahigh field for multiparametric stroke assessment, are discussed.


Assuntos
Imagem de Difusão por Ressonância Magnética , Compostos Férricos/química , AVC Isquêmico/diagnóstico por imagem , Células-Tronco Mesenquimais/metabolismo , Tamanho da Partícula , Sódio/química , Acidente Vascular Cerebral/diagnóstico por imagem , Animais , Biomarcadores/metabolismo , Humanos , Infarto da Artéria Cerebral Média/patologia , Curva ROC , Ratos
11.
Cytotherapy ; 21(10): 1033-1048, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31537468

RESUMO

Human mesenchymal stem cells (hMSCs) have been shown to enhance stroke lesion recovery by mediating inflammation and tissue repair through secretion of trophic factors. However, low cell survival and reduced primitive stem cell function of culture-expanded hMSCs are the major challenges limiting hMSC therapeutic efficacy in stroke treatment. In this study, we report the effects of short-term preconditioning of hMSCs via three-dimensional (3D) aggregation on stroke lesion recovery after intra-arterial (IA) transplantation of 3D aggregate-derived hMSCs (Agg-D hMSCs) in a transient middle cerebral artery occlusion (MCAO) stroke model. Compared with two-dimensional (2D) monolayer culture, Agg-D hMSCs exhibited increased resistance to ischemic stress, secretory function and therapeutic outcome. Short-term preconditioning via 3D aggregation reconfigured hMSC energy metabolism and altered redox cycle, which activated the PI3K/AKT pathway and enhanced resistance to in vitro oxidative stress. Analysis of transplanted hMSCs in MCAO rats using ultra-high-field magnetic resonance imaging at 21.1 T showed increased hMSC persistence and stroke lesion reduction by sodium (23Na) imaging in the Agg-D hMSC group compared with 2D hMSC control. Behavioral analyses further revealed functional improvement in MCAO animal treated with Agg-D hMSCs compared with saline control. Together, the results demonstrated the improved outcome for Agg-D hMSCs in the MCAO model and suggest short-term 3D aggregation as an effective preconditioning strategy for hMSC functional enhancement in stroke treatment.


Assuntos
Sobrevivência de Enxerto/fisiologia , Infarto da Artéria Cerebral Média/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Acidente Vascular Cerebral/terapia , Adulto , Animais , Agregação Celular/fisiologia , Células Cultivadas , Humanos , Infarto da Artéria Cerebral Média/patologia , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia , Resultado do Tratamento , Adulto Jovem
12.
PLoS One ; 14(6): e0218041, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31173612

RESUMO

There is strong evidence that neuronal hyper-excitability underlies migraine, and may or may not be preceded by cortical spreading depression. However, the mechanisms for cortical spreading depression and/or migraine are not established. Previous studies reported that cerebrospinal fluid (CSF) [Na+] is higher during migraine, and that higher extracellular [Na+] leads to hyper-excitability. We raise the hypothesis that altered choroid plexus Na+, K+-ATPase activity can cause both migraine phenomena: inhibition raises CSF [K+] and initiates cortical spreading depression, while activation raises CSF [Na+] and causes migraine. In this study, we examined levels of specific Na+, K+-ATPase inhibitors, endogenous ouabain-like compounds (EOLC), in CSF from migraineurs and controls. CSF EOLC levels were significantly lower during ictal migraine (0.4 nM +/- 0.09) than from either controls (1.8 nM +/- 0.4) or interictal migraineurs (3.1 nM +/- 1.9). Blood plasma EOLC levels were higher in migraineurs than controls, but did not differ between ictal and interictal states. In a Sprague-Dawley rat model of nitroglycerin-triggered central sensitization, we changed the concentrations of EOLC and CSF sodium, and measured aversive mechanical threshold (von Frey hairs), trigeminal nucleus caudalis activation (cFos), and CSF [Na+] (ultra-high field 23Na MRI). Animals were sensitized by three independent treatments: intraperitoneal nitroglycerin, immunodepleting EOLC from cerebral ventricles, or cerebroventricular infusion of higher CSF [Na+]. Conversely, nitroglycerin-triggered sensitization was prevented by either vascular or cerebroventricular delivery of the specific Na+, K+-ATPase inhibitor, ouabain. These results affirm our hypothesis that higher CSF [Na+] is linked to human migraine and to a rodent migraine model, and demonstrate that EOLC regulates them both. Our data suggest that altered choroid plexus Na+, K+-ATPase activity is a common source of these changes, and may be the initiating mechanism in migraine.


Assuntos
Líquido Cefalorraquidiano/metabolismo , Íons/metabolismo , Transtornos de Enxaqueca/etiologia , Transtornos de Enxaqueca/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sódio/metabolismo , Adolescente , Adulto , Idoso , Animais , Plexo Corióideo/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ouabaína/metabolismo , Ratos , Ratos Sprague-Dawley , Adulto Jovem
13.
Brain Circ ; 4(3): 133-138, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30450421

RESUMO

Ischemia, which involves decreased blood flow to a region and a corresponding deprivation of oxygen and nutrients, can be induced as a consequence of stroke or heart attack. A prevalent disease that affects many individuals worldwide, ischemic stroke results in functional and cognitive impairments, as neural cells in the brain receive inadequate nourishment and encounter inflammation and various other detrimental toxic factors that lead to their death. Given the scarce treatments for this disease in the clinic such as the administration of tissue plasminogen activator, which is only effective in a limited time window after the occurrence of stroke, it will be necessary to develop new strategies to ameliorate or prevent stroke-induced brain damage. Cell-based therapies appear to be a promising solution for treating ischemic stroke and many other ischemia-associated and neurodegenerative maladies. Particularly, human mesenchymal stem cells (hMSCs) are of interest for cell transplantation in stroke, given their multipotency, accessibility, and reparative abilities. To determine the fate and survival of hMSC, which will be imperative for successful transplantation therapies, these cells may be monitored using magnetic resonance imaging and transfected with superparamagnetic iron oxide (SPIO), a contrast agent that facilitates the detection of these hMSCs. This review encompasses pertinent research and findings to reveal the effects of SPIO on hMSC functions in the context of transplantation in ischemic environments and over extended time periods. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.

14.
NMR Biomed ; 31(11): e3995, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30052292

RESUMO

This study explores opportunities opened up by ultrahigh fields for in vivo saturation transfer brain magnetic resonance imaging experiments. Fast spin-echo images weighted by chemical exchange saturation transfer (CEST) effects were collected on Sprague-Dawley rats at 21.1 T, focusing on two neurological models. One involved a middle cerebral artery occlusion emulating ischemic stroke; the other involved xenografted glioma cells that were followed over the course of several days as they developed into brain tumors. A remarkably strong saturation-derived contrast was observed for the growing tumors when calculating magnetization transfer ratios at c. 3.8 ppm. This large contrast originated partially from an increase in the contribution of the amide CEST effect, but mostly from strong decreases in the Overhauser and magnetization transfer contributions to the upfield region, whose differential attenuations could be clearly discerned thanks to the ultrahigh field. The high spectral separation arising at 21.1 T also revealed numerous CEST signals usually overlapping at lower fields. Ischemic lesions were also investigated but, remarkably, magnetization and saturation transfer contrasts were nearly absent when computing transfer asymmetries using either high or low saturation power schemes. These behaviors were consistently observed at 24 hours post-occlusion, regardless of the data processing approach assayed. Considerations related to how various parameters defining these experiments depend on the magnetic field, primarily chemical shifts and T1 values, are discussed.


Assuntos
Encéfalo/diagnóstico por imagem , Imageamento por Ressonância Magnética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioma/diagnóstico por imagem , Glioma/patologia , Ratos Sprague-Dawley
15.
Sci Rep ; 8(1): 2710, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29426863

RESUMO

Cocaine is one of the powerful addictive drugs, widely abused in most Western countries. Because of high lipophilic nature, cocaine easily reaches various domains of the central nervous system (CNS) and triggers different levels of cellular toxicity. The aim of this investigation was to reproduce cocaine toxicity in differentiated PC12 cells through quantitative knowledge on biochemical and cytotoxicity markers. We differentiated the cells with 0.1 µg/ml nerve growth factor (NGF) for 5 days, followed by treatment with cocaine for 48 h at in vivo and in vitro concentrations. Results indicated that cocaine at in vivo concentrations neither killed the cells nor altered the morphology, but decreased the mitochondrial membrane potential that paralleled with increased lactate and glutathione (GSH) levels. On the other hand, cocaine at in vitro concentrations damaged the neurites and caused cell death, which corresponded with increased reactive oxygen species (ROS) generation, plasma membrane damage, and GSH depletion with no detectable nitric oxide (NO) level. While direct understanding of cocaine and cell interaction under in vivo animal models is impeded due to high complexity, our present in vitro results assisted in understanding the onset of some key events of neurodegenerative diseases in cocaine treated neuronal cells.


Assuntos
Cocaína/toxicidade , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Sobrevivência Celular , Glutationa/metabolismo , Ácido Láctico/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Fator de Crescimento Neural/química , Neuritos/metabolismo , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo
16.
Restor Neurol Neurosci ; 34(3): 433-41, 2016 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-27080073

RESUMO

PURPOSE: Major depression and related mood disorders are the most common long-term outcomes associated with traumatic brain injury (TBI). Given the potentially debilitating consequences of depression, and the fact that TBI patients are frequently refractory to antidepressant drugs, new therapies are clearly needed. We hypothesized that human bone marrow-derived mesenchymal stem cells (hMSC), delivered intravenously, can effectively treat TBI-induced depression and other behavioral deficits associated with TBI. METHODS: Rats (n = 8 per group) were subjected to experimental TBI or control sham operation. Six hours post TBI, rats were treated with 1×106 hMSC or vehicle control. Immediately after TBI and prior to hMSC or control treatment, rats were subjected to either targeted precision x-ray irradiation to eliminate subventricular zone (SVZ) proliferation or sham irradiation. One week after TBI, SVZ irradiation, and hMSC treatment, rats were evaluated for the depression-like behavior, anhedonia, using the two-bottle saccharin preference paradigm; and for working memory using the novel object recognition test. RESULTS: TBI resulted in a 54% (p≤0.05) decrease in saccharin preference scores while treatment of TBI with hMSC fully prevented this anhedonic behavior. TBI was also found to produce a 73% (p≤0.05) decrease in novel object interaction time, indicating impaired working memory, and was similarly improved by treatment with hMSC. The ability of hMSC to prevent TBI-associated depression and working memory impairment was eliminated when SVZ proliferation was inhibited by irradiation. CONCLUSIONS: This work has identified a possible role for hMSC in the treatment of TBI-induced depression and other behaviors and suggests a mechanistic role for proliferative cells of the SVZ proliferation in hMSC efficacy.


Assuntos
Anedonia/fisiologia , Lesões Encefálicas Traumáticas/psicologia , Lesões Encefálicas Traumáticas/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/etiologia , Proliferação de Células/fisiologia , Comportamento Exploratório , Humanos , Comportamento Impulsivo/fisiologia , Ventrículos Laterais/citologia , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/fisiologia , Lesões por Radiação/complicações , Lesões por Radiação/terapia , Ratos , Ratos Sprague-Dawley , Reconhecimento Psicológico , Fatores de Tempo
17.
Metabolism ; 65(5): 685-698, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27085776

RESUMO

PURPOSE: The appropriate mode of exercise training for cancer cachexia is not well-established. Using the colon-26 (C26) mouse model of cancer cachexia, we defined and compared the skeletal muscle responses to aerobic and resistance training. METHODS: Twelve-month old Balb/c mice were initially assigned to control, aerobic training (AT; wheel running), or resistance training (RT; ladder climbing) (n=16-17/group). After 8weeks of training, half of each group was injected with C26 tumor cells, followed by 3 additional weeks of training. Body composition and neuromuscular function was evaluated pre- and post-training. Muscles were collected post-training and analyzed for fiber cross-sectional area (CSA), Akt-mTOR signaling, and expression of insulin-like growth factor-I (IGF-I) and myogenic regulatory factors. RESULTS: Total body mass decreased (p<0.05) in C26 (-8%), AT+C26 (-18%), and RT+C26 (-15%) but not control. Sensorimotor function declined (p<0.05) in control (-16%), C26 (-13%), and RT+C26 (-23%) but not AT+C26. Similarly, strength/body weight decreased (p<0.05) in control (-7%), C26 (-21%), and RT+C26 (-10%) but not AT+C26. Gastrocnemius mass/body weight tended to be greater in AT+C26 vs. C26 (+6%, p=0.09). Enlargement of the spleen was partially corrected in AT+C26 (-27% vs. C26, p<0.05). Fiber CSA was lower in all C26 groups vs. control (-32% to 46%, p<0.05); however, the effect size calculated from C26 and AT+C26 was large (+24%, d=1.04). Phosphorylated levels of mTOR in AT+C26 exceeded C26 (+32%, p<0.05). RT+C26 showed greater mRNA expression (p<0.05) of IGF-IEa (+79%) and myogenin (+126%) with a strong tendency for greater IGF-IEb (+127%, p=0.069) vs. CONCLUSIONS: Aerobic or resistance training was unable to prevent tumor-induced body weight loss. However, aerobic training may have preserved function, reduced the inflammatory response of the spleen, and marginally rescued muscle mass possibly through activation of mTOR. Aerobic training may therefore have therapeutic value for patients with cancer cachexia. In contrast, resistance training induced the expression of genes associated with muscle damage and repair. This gene response may be supportive of excessive stress generated by high resistance loading in a tumor-bearing state.


Assuntos
Caquexia/prevenção & controle , Neoplasias do Colo/terapia , Modelos Animais de Doenças , Atividade Motora , Desenvolvimento Muscular , Músculo Esquelético/patologia , Treinamento Resistido , Animais , Caquexia/etiologia , Caquexia/fisiopatologia , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Feminino , Regulação Neoplásica da Expressão Gênica , Camundongos Endogâmicos BALB C , Debilidade Muscular/etiologia , Debilidade Muscular/prevenção & controle , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Transplante de Neoplasias , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/agonistas , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Transdução de Sinais , Serina-Treonina Quinases TOR/química , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
18.
Biotechnol Prog ; 31(2): 510-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25905549

RESUMO

Magnetic resonance imaging (MRI) provides an effective approach to track labeled pluripotent stem cell (PSC)-derived neural progenitor cells (NPCs) for neurological disorder treatments after cell labeling with a contrast agent, such as an iron oxide derivative. Cryopreservation of pre-labeled neural cells, especially in three-dimensional (3D) structure, can provide a uniform cell population and preserve the stem cell niche for the subsequent applications. In this study, the effects of cryopreservation on PSC-derived multicellular NPC aggregates labeled with micron-sized particles of iron oxide (MPIO) were investigated. These NPC aggregates were labeled prior to cryopreservation because labeling thawed cells can be limited by inefficient intracellular uptake, variations in labeling efficiency, and increased culture time before use, minimizing their translation to clinical settings. The results indicated that intracellular MPIO incorporation was retained after cryopreservation (70-80% labeling efficiency), and MPIO labeling had little adverse effects on cell recovery, proliferation, cytotoxicity and neural lineage commitment post-cryopreservation. MRI analysis showed comparable detectability for the MPIO-labeled cells before and after cryopreservation indicated by T2 and T2* relaxation rates. Cryopreserving MPIO-labeled 3D multicellular NPC aggregates can be applied in in vivo cell tracking studies and lead to more rapid translation from preservation to clinical implementation.


Assuntos
Criopreservação , Células-Tronco Embrionárias/citologia , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita/química , Células-Tronco Neurais/citologia , Animais , Agregação Celular , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Camundongos , Células-Tronco Neurais/fisiologia , Espécies Reativas de Oxigênio/metabolismo
19.
Methods Mol Biol ; 1283: 43-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25304204

RESUMO

Due to the unlimited proliferation capacity and the unique differentiation ability of pluripotent stem cells (PSCs), including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), large numbers of PSC-derived cell products are in demand for applications in drug screening, disease modeling, and especially cell therapy. In stem cell-based therapy, tracking transplanted cells with magnetic resonance imaging (MRI) has emerged as a powerful technique to reveal cell survival and distribution. This chapter illustrated the basic steps of labeling PSC-derived neural progenitors (NPs) with micron-sized particles of iron oxide (MPIO, 0.86 µm) for MRI analysis. The protocol described PSC expansion and differentiation into NPs, and the labeling of the derived cells either after replating on adherent surface or in suspension. The labeled cells can be analyzed using in vitro MRI analysis. The methods presented here can be easily adapted for cell labeling in cell processing facilities under current Good Manufacturing Practices (cGMP). The iron oxide-labeled NPs can be used for cellular monitoring of in vitro cultures and in vivo transplantation.


Assuntos
Diferenciação Celular , Rastreamento de Células/métodos , Compostos Férricos/metabolismo , Imageamento por Ressonância Magnética , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos
20.
Cytotherapy ; 17(1): 98-111, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25527864

RESUMO

BACKGROUND AIMS: Pluripotent stem cell (PSC)-derived neural progenitor cells (NPCs) represent an unlimited source for the treatment of various neurological disorders. NPCs are usually derived from PSCs through the formation of embryoid body (EB), an aggregate structure mimicking embryonic development. This study investigated the effect of labeling multicellular EB-NPC aggregates with micron-sized particles of iron oxide (MPIO) for cell tracking using magnetic resonance imaging (MRI). METHODS: Intact and dissociated EB-NPC aggregates were labeled with various concentrations of MPIOs (0, 2.5, 5 and 10 µg Fe/mL). The labeled cells were analyzed by fluorescent imaging, flow cytometry and in vitro MRI for labeling efficiency and detectability. Moreover, the biological effects of intracellular MPIO on cell viability, cytotoxicity, proliferation and neural differentiation were evaluated. RESULTS: Intact EB-NPC aggregates showed higher cell proliferation and viability compared with the dissociated cells. Despite diffusion limitation at low MPIO concentration, higher concentration of MPIO (i.e., 10 µg Fe/mL) was able to label EB-NPC aggregates at similar efficiency to the single cells. In vitro MRI showed concentration-dependent MPIO detection in EB-NPCs over 2.0-2.6 population doublings. More important, MPIO incorporation did not affect the proliferation and neural differentiation of EB-NPCs. CONCLUSIONS: Multicellular EB-NPC aggregates can be efficiently labeled and tracked with MPIO while maintaining cell proliferation, phenotype and neural differentiation potential. This study demonstrated the feasibility of labeling EB-NPC aggregates with MPIO for cellular monitoring of in vitro cultures and in vivo transplantation.


Assuntos
Células-Tronco Embrionárias/citologia , Compostos Férricos/farmacologia , Nanopartículas de Magnetita/administração & dosagem , Células-Tronco Mesenquimais/citologia , Células-Tronco Neurais/citologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Rastreamento de Células/métodos , Células-Tronco Embrionárias/efeitos dos fármacos , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Tamanho da Partícula , Coloração e Rotulagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA