Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
NMR Biomed ; 32(2): e4037, 2019 02.
Article in English | MEDLINE | ID: mdl-30489666

ABSTRACT

Alzheimer's disease is the most common neurodegenerative disease, and many patients also present with vascular dysfunction. In this study, we aimed to assess cerebral blood flow (CBF) and cerebrovascular response (CVR) as early, pre-symptomatic (3 months of age), imaging markers in a bigenic model of Alzheimer's disease (APP.V717IxTau.P301L, biAT) and in the monogenic parental strains. We further developed our previously published combination of pulsed arterial spin labeling perfusion MRI and hypo-ventilation paradigm, which allows weaning of the mice from the ventilator. Furthermore, the commonly used isoflurane anesthesia induces vasodilation and is thereby inherently a vascular challenge. We therefore assessed perfusion differences in the mouse models under free-breathing isoflurane conditions. We report (i) that we can determine CBF and hypoventilation-based CVR under ketamine/midazolam anesthesia and wean mice from the ventilator, making it a valuable tool for assessment of CBF and CVR in mice, (ii) that biAT mice exhibit lower cortical CBF than wild-type mice at age 3 months, (iii) that CVR was increased in both biAT and APP.V717I mice but not in Tau.P301L mice, identifying the APP genotype as a strong influencer of brain CVR and (iv) that perfusion differences at baseline are masked by the widely used isoflurane anesthesia.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Protein Precursor/metabolism , Brain/blood supply , Hypoventilation/complications , Hypoventilation/physiopathology , Perfusion , tau Proteins/metabolism , Anesthesia , Animals , Carbon Dioxide/metabolism , Disease Models, Animal , Isoflurane/administration & dosage , Isoflurane/pharmacology , Male , Mice, Transgenic , Plaque, Amyloid/pathology
2.
Cytotherapy ; 19(6): 744-755, 2017 06.
Article in English | MEDLINE | ID: mdl-28499585

ABSTRACT

BACKGROUND AIMS: Myelodysplastic syndromes (MDS) are a group of clonal stem cell disorders affecting the normal hematopoietic differentiation process and leading to abnormal maturation and differentiation of all blood cell lineages. Treatment options are limited, and there is an unmet medical need for effective therapies for patients with severe cytopenias. METHODS: We demonstrate that multipotent adult progenitor cells (MAPC) improve the function of hematopoietic progenitors derived from human MDS bone marrow (BM) by significantly increasing the frequency of primitive progenitors as well as the number of myeloid colonies. RESULTS: This effect was more pronounced in a non-contact culture, indicating the importance of soluble factors produced by the MAPC cells. Moreover, the cells did not stimulate the growth of the abnormal MDS clone, as shown by fluorescent in situ hybridization analysis on BM cells from patients with a known genetic abnormality. We also demonstrate that MAPC cells can provide stromal support for patient-derived hematopoietic cells. When MAPC cells were intravenously injected into a mouse model of MDS, they migrated to the site of injury and increased the hematopoietic function in diseased mice. DISCUSSION: The preclinical studies undertaken here indicate an initial proof of concept for the use of MAPC cell therapy in patients with MDS-related severe and symptomatic cytopenias and should pave the way for further investigation in clinical trials.


Subject(s)
Multipotent Stem Cells/transplantation , Myelodysplastic Syndromes/therapy , Adult , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Female , Hematopoiesis , Humans , In Situ Hybridization, Fluorescence , Mice, Inbred C57BL
3.
Nanomedicine ; 13(5): 1663-1671, 2017 07.
Article in English | MEDLINE | ID: mdl-28366819

ABSTRACT

A major conceptual breakthrough in cell signaling has been the finding of EV as new biomarker shuttles in body fluids. Now, one of the major challenges in using these nanometer-sized biological entities as diagnostic marker is the development of translational methodologies to profile them. SPR offers a promising label-free and real time platform with a high potential for biomarker detection. Therefore, we aimed to develop a uniform SPR methodology to detect specific surface markers on EV derived from patient with CHD. EVs having an approximate size range between 30 and 100 nm (~48.5%) and 100-300 nm (~51.5%) were successfully isolated. The biomarker profile of EV was verified using immunogold labeling, ELISA and SPR. Using SPR, we demonstrated an increased binding of EV derived from patients with CHD to anti-ICAM-1 antibodies as compared to EV from healthy donors. Our current findings open up novel opportunities for in-depth and label-free investigation of EV.


Subject(s)
Biomarkers , Endothelial Cells , Extracellular Vesicles , Surface Plasmon Resonance , Coronary Disease , Humans , Inflammation , Nanotechnology/methods
4.
Med Res Rev ; 36(6): 1080-1126, 2016 11.
Article in English | MEDLINE | ID: mdl-27439773

ABSTRACT

Stroke is the second most common cause of death and is a major cause of permanent disability. Given the current demographic trend of an ageing population and associated increased risk, the prevalence of and socioeconomic burden caused by stroke will continue to rise. Current therapies are unable to sufficiently ameliorate the disease outcome and are not applicable to all patients. Therefore, strategies such as cell-based therapies with mesenchymal stem cell (MSC) or induced pluripotent stem cell (iPSC) pave the way for new treatment options for stroke. These cells showed great preclinical promise despite the fact that the precise mechanism of action and the optimal administration route are unknown. To gain dynamic insights into the underlying repair processes after stem cell engraftment, noninvasive imaging modalities were developed to provide detailed spatial and functional information on the donor cell fate and host microenvironment. This review will focus on MSCs and iPSCs as types of widely used stem cell sources in current (bio)medical research and compare their efficacy and potential to ameliorate the disease outcome in animal stroke models. In addition, novel noninvasive imaging strategies allowing temporospatial in vivo tracking of transplanted cells and coinciding evaluation of neuronal repair following stroke will be discussed.


Subject(s)
Brain Ischemia/therapy , Stem Cell Transplantation/methods , Stroke/therapy , Animals , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Humans , Induced Pluripotent Stem Cells/transplantation , Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation/methods , Positron-Emission Tomography/methods , Regeneration/physiology , Stroke/diagnostic imaging , Stroke/pathology , Tomography, Emission-Computed, Single-Photon/methods
5.
Stem Cells ; 32(11): 2833-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142614

ABSTRACT

Adult stem cells have been investigated increasingly over the past years for multiple applications. Although they have a more favorable safety profile compared to pluripotent stem cells, they are still capable of self-renewal and differentiate into several cell types. We investigated the behavior of Oct4-positive (Oct4(+)) and Oct4-negative (Oct4(-) ) murine or rat bone marrow (BM)-derived stem cells in the healthy brain of syngeneic mice and rats. Engraftment of mouse and rat Oct4-positive BM-derived hypoblast-like stem cells (m/rOct4(+) BM-HypoSCs) resulted in yolk-sac tumor formation in the healthy brain which was monitored longitudinally using magnetic resonance imaging (MRI) and bioluminescence imaging (BLI). Contrast enhanced MRI confirmed the disruption of the blood brain barrier. In contrast, m/r Oct4-negative BM-derived multipotent adult progenitor cells (m/rOct4(-) BM-MAPCs) did not result in mass formation after engraftment into the brain. mOct4(+) BM-HypoSCs and mOct4(-) BM-MAPCs were transduced to express enhanced green fluorescent protein, firefly luciferase (fLuc), and herpes simplex virus-thymidine kinase to follow up suicide gene expression as a potential "safety switch" for tumor-forming stem cells by multimodal imaging. Both cell lines were eradicated efficiently in vivo by ganciclovir administration indicating successful suicide gene expression in vivo, as assessed by MRI, BLI, and histology. The use of suicide genes to prevent tumor formation is in particular of interest for therapeutic approaches where stem cells are used as vehicles to deliver therapeutic genes.


Subject(s)
Ganciclovir/metabolism , Stem Cells/cytology , Animals , Brain/metabolism , Cell Line , Green Fluorescent Proteins/metabolism , Luciferases, Firefly/metabolism , Magnetic Resonance Imaging , Mice , Models, Animal , Octamer Transcription Factor-3/metabolism , Rabbits , Rats
6.
FASEB J ; 28(4): 1634-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24352035

ABSTRACT

In the present study, we evaluated the differentiation potential of human dental pulp stem cells (hDPSCs) toward Schwann cells, together with their functional capacity with regard to myelination and support of neurite outgrowth in vitro. Successful Schwann cell differentiation was confirmed at the morphological and ultrastructural level by transmission electron microscopy. Furthermore, compared to undifferentiated hDPSCs, immunocytochemistry and ELISA tests revealed increased glial marker expression and neurotrophic factor secretion of differentiated hDPSCs (d-hDPSCs), which promoted survival and neurite outgrowth in 2-dimensional dorsal root ganglia cultures. In addition, neurites were myelinated by d-hDPSCs in a 3-dimensional collagen type I hydrogel neural tissue construct. This engineered construct contained aligned columns of d-hDPSCs that supported and guided neurite outgrowth. Taken together, these findings provide the first evidence that hDPSCs are able to undergo Schwann cell differentiation and support neural outgrowth in vitro, proposing them to be good candidates for cell-based therapies as treatment for peripheral nerve injury.


Subject(s)
Cell Differentiation , Dental Pulp/cytology , Neurites/physiology , Schwann Cells/cytology , Stem Cells/cytology , Tissue Engineering/methods , Adolescent , Animals , Animals, Newborn , Cell Culture Techniques/methods , Cells, Cultured , Collagen/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , Laminin/metabolism , Microscopy, Electron, Transmission , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nestin/metabolism , Neurites/metabolism , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Schwann Cells/ultrastructure , Stem Cells/metabolism , Young Adult
7.
J Neurooncol ; 119(2): 297-306, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990826

ABSTRACT

Blood brain barrier (BBB) disruption is used (pre)clinically as a measure for brain tumor malignancy and grading. During treatment it is one of the parameters followed rigorously to assess therapeutic efficacy. In animal models, both invasive and non-invasive methods are used to determine BBB disruption, among them Evans blue injection prior to sacrifice and T1-weighted magnetic resonance imaging (MRI) post contrast injection. In this study, we have assessed the BBB integrity with the methods mentioned above in two experimental high grade glioma models, namely the GL261 mouse glioblastoma model and the Hs683 human oligodendroglioma model. The GL261 model showed clear BBB integrity loss with both, contrast-enhanced (CE) MRI and Evans blue staining. In contrast, the Hs683 model only displayed BBB disruption with CE-MRI, which was not evident on Evans blue staining, indicating a limited BBB disruption. These results clearly indicate the importance of assessing the BBB integrity status using appropriate methods. Especially when using large therapeutic molecules that have difficulties crossing the BBB, care should be taken with the appropriate BBB disruption assessment studies.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Capillary Permeability/physiology , Glioblastoma/metabolism , Oligodendroglioma/metabolism , Animals , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Evans Blue , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Mice, Inbred C57BL , Neoplasm Grading , Oligodendroglioma/pathology
8.
Mediators Inflamm ; 2013: 685317, 2013.
Article in English | MEDLINE | ID: mdl-23818742

ABSTRACT

Demyelination is one of the pathological hallmarks of multiple sclerosis (MS). To date, no therapy is available which directly potentiates endogenous remyelination. Interleukin-11 (IL-11), a member of the gp130 family of cytokines, is upregulated in MS lesions. Systemic IL-11 treatment was shown to ameliorate clinical symptoms in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. IL-11 modulates immune cells and protects oligodendrocytes in vitro. In this study, the cuprizone-induced demyelination mouse model was used to elucidate effects of IL-11 on de- and remyelination, independent of the immune response. Prophylactic-lentiviral- (LV-) mediated overexpression of IL-11 in mouse brain significantly limited acute demyelination, which was accompanied with the preservation of CC1(+) mature oligodendrocytes (OLs) and a decrease in microglial activation (Mac-2(+)). We further demonstrated that IL-11 directly reduces myelin phagocytosis in vitro. When IL-11 expressing LV was therapeutically applied in animals with extensive demyelination, a significant enhancement of remyelination was observed as demonstrated by Luxol Fast Blue staining and electron microscopy imaging. Our results indicate that IL-11 promotes maturation of NG2(+) OPCs into myelinating CC1(+) OLs and may thus explain the enhanced remyelination. Overall, we demonstrate that IL-11 is of therapeutic interest for MS and other demyelinating diseases by limiting demyelination and promoting remyelination.


Subject(s)
Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Interleukin-11/metabolism , Animals , Cell Proliferation/drug effects , Central Nervous System/ultrastructure , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , Humans , Immunohistochemistry , Interleukin-11/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Microglia/ultrastructure , Microscopy, Electron, Transmission , Myelin Sheath/metabolism
9.
Eur J Nucl Med Mol Imaging ; 39(11): 1796-806, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22895861

ABSTRACT

PURPOSE: Recent ex vivo and pharmacological evidence suggests involvement of the endocannabinoid system in the pathophysiology of stroke, but conflicting roles for type 1 and 2 cannabinoid receptors (CB(1) and CB(2)) have been suggested. The purpose of this study was to evaluate CB(1) and CB(2) receptor binding over time in vivo in a rat photothrombotic stroke model using PET. METHODS: CB(1) and CB(2) microPET imaging was performed at regular time-points up to 2 weeks after stroke using [(18)F]MK-9470 and [(11)C]NE40. Stroke size was measured using MRI at 9.4 T. Ex vivo validation was performed via immunostaining for CB(1) and CB(2). Immunofluorescent double stainings were also performed with markers for astrocytes (GFAP) and macrophages/microglia (CD68). RESULTS: [(18)F]MK-9470 PET showed a strong increase in CB(1) binding 24 h and 72 h after stroke in the cortex surrounding the lesion, extending to the insular cortex 24 h after surgery. These alterations were consistently confirmed by CB(1) immunohistochemical staining. [(11)C]NE40 did not show any significant differences between stroke and sham-operated animals, although staining for CB(2) revealed minor immunoreactivity at 1 and 2 weeks after stroke in this model. Both CB (1) (+) and CB (2) (+) cells showed minor immunoreactivity for CD68. CONCLUSION: Time-dependent and regionally strongly increased CB(1), but not CB(2), binding are early consequences of photothrombotic stroke. Pharmacological interventions should primarily aim at CB(1) signalling as the role of CB(2) seems minor in the acute and subacute phases of stroke.


Subject(s)
Positron-Emission Tomography , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Stroke/diagnostic imaging , Animals , Disease Models, Animal , Pyridines , Quinolines , Rats , Stroke/etiology , Stroke/metabolism , Thrombosis/complications
10.
Pharmaceutics ; 13(9)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34575570

ABSTRACT

The use of multimodal contrast agents can potentially overcome the intrinsic limitations of individual imaging methods. We have validated synthetic antiferromagnetic nanoparticles (SAF-NPs) as bimodal contrast agents for in vitro cell labeling and in vivo cell tracking using magnetic resonance imaging (MRI) and computed tomography (CT). SAF-NP-labeled cells showed high contrast in MRI phantom studies (r2* = 712 s-1 mM-1), while pelleted cells showed clear contrast enhancement in CT. After intravenous SAF-NP injection, nanoparticles accumulated in the liver and spleen, as visualized in vivo by significant MRI contrast enhancement. Intravenous injection of SAF-NP-labeled cells resulted in cell accumulation in the lungs, which was clearly detectable by using CT but not by using MRI. SAF-NPs proved to be very efficient cell labeling agents for complementary MRI- and CT-based cell tracking. Bimodal monitoring of SAF-NP labeled cells is in particular of interest for applications where the applied imaging methods are not able to visualize the particles and/or cells in all organs.

11.
Nanomaterials (Basel) ; 10(11)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33114177

ABSTRACT

Gold nanoparticles offer the possibility to combine both imaging and therapy of otherwise difficult to treat tumors. To validate and further improve their potential, we describe the use of gold nanostars that were functionalized with a polyethyleneglycol-maleimide coating for in vitro and in vivo photoacoustic imaging (PAI), computed tomography (CT), as well as photothermal therapy (PTT) of cancer cells and tumor masses, respectively. Nanostar shaped particles show a high absorption coefficient in the near infrared region and have a hydrodynamic size in biological medium around 100 nm, which allows optimal intra-tumoral retention. Using these nanostars for in vitro labeling of tumor cells, high intracellular nanostar concentrations could be achieved, resulting in high PAI and CT contrast and effective PTT. By injecting the nanostars intratumorally, high contrast could be generated in vivo using PAI and CT, which allowed successful multi-modal tumor imaging. PTT was successfully induced, resulting in tumor cell death and subsequent inhibition of tumor growth. Therefore, gold nanostars are versatile theranostic agents for tumor therapy.

12.
J Pers Med ; 8(1)2018 Mar 12.
Article in English | MEDLINE | ID: mdl-29534555

ABSTRACT

Pancreatic islets (PIs) transplantation is an alternative approach for the treatment of severe forms of type 1 diabetes (T1D). To monitor the success of transplantation, it is desirable to follow the location of engrafted PIs non-invasively. In vivo magnetic resonance imaging (MRI) of transplanted PIs is a feasible cell tracking method; however, this requires labeling with a suitable contrast agent prior to transplantation. We have tested the feasibility of cationic magnetoliposomes (MLs), compared to commercial contrast agents (Endorem and Resovist), by labeling insulinoma cells and freshly isolated rat PIs. It was possible to incorporate Magnetic Ressonance (MR)-detectable amounts of MLs in a shorter time (4 h) when compared to Endorem and Resovist. MLs did not show negative effects on the PIs' viability and functional parameters in vitro. Labeled islets were transplanted in the renal sub-capsular region of healthy mice. Hypointense contrast in MR images due to the labeled PIs was detected in vivo upon transplantation, while MR detection of PIs labeled with Endorem and Resovist was only possible after the addition of transfection agents. These findings indicate that MLs are suitable to image PIs, without affecting their function, which is promising for future longitudinal pre-clinical and clinical studies involving the assessment of PI transplantation.

13.
Sci Rep ; 8(1): 11487, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30065302

ABSTRACT

Magnetoliposomes (MLs) were synthesized and tested for longitudinal monitoring of transplanted pancreatic islets using magnetic resonance imaging (MRI) in rat models. The rat insulinoma cell line INS-1E and isolated pancreatic islets from outbred and inbred rats were used to optimize labeling conditions in vitro. Strong MRI contrast was generated by islets exposed to 50 µg Fe/ml for 24 hours without any increased cell death, loss of function or other signs of toxicity. In vivo experiments showed that pancreatic islets (50-1000 units) labeled with MLs were detectable for up to 6 weeks post-transplantation in the kidney subcapsular space. Islets were also monitored for two weeks following transplantation through the portal vein of the liver. Hereby, islets labeled with MLs and transplanted under the left kidney capsule were able to correct hyperglycemia and had stable MRI signals until nephrectomy. Interestingly, in vivo MRI of streptozotocin induced diabetic rats transplanted with allogeneic islets demonstrated loss of MRI contrast between 7-16 days, indicative of loss of islet structure. MLs used in this study were not only beneficial for monitoring the location of transplanted islets in vivo with high sensitivity but also reported on islet integrity and hereby indirectly on islet function and rejection.


Subject(s)
Contrast Media/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Magnetite Nanoparticles/administration & dosage , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin/metabolism , Islets of Langerhans Transplantation/methods , Liver/metabolism , Liver/pathology , Longitudinal Studies , Magnetic Resonance Imaging/methods , Portal Vein/metabolism , Portal Vein/pathology , Rats , Rats, Inbred Lew , Rats, Wistar , Streptozocin/pharmacology
14.
J Cereb Blood Flow Metab ; 37(2): 726-739, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26994041

ABSTRACT

We have characterized both acute and long-term vascular and metabolic effects of unilateral common carotid artery occlusion in mice by in vivo magnetic resonance imaging and positron emission tomography. This common carotid artery occlusion model induces chronic cerebral hypoperfusion and is therefore relevant to both preclinical stroke studies, where it serves as a control condition for a commonly used mouse model of ischemic stroke, and neurodegeneration, as chronic hypoperfusion is causative to cognitive decline. By using perfusion magnetic resonance imaging, we demonstrate that under isoflurane anesthesia, cerebral perfusion levels recover gradually over one month. This recovery is paralleled by an increase in lumen diameter and altered tortuosity of the contralateral internal carotid artery at one year post-ligation as derived from magnetic resonance angiography data. Under urethane/α-chloralose anesthesia, no acute perfusion differences are observed, but the vascular response capacity to hypercapnia is found to be compromised. These hemispheric perfusion alterations are confirmed by water [15O]-H2O positron emission tomography. Glucose metabolism ([18F]-FDG positron emission tomography) or white matter organization (diffusion-weighted magnetic resonance imaging) did not show any significant alterations. In conclusion, permanent unilateral common carotid artery occlusion results in acute and long-term vascular remodeling, which may have immediate consequences for animal models of stroke but also vascular dementia.


Subject(s)
Brain Ischemia/pathology , Brain/blood supply , Brain/pathology , Carotid Artery, Common/pathology , Cerebrovascular Circulation , Vascular Remodeling , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Ischemia/diagnostic imaging , Brain Ischemia/metabolism , Carotid Artery, Common/diagnostic imaging , Carotid Artery, Common/metabolism , Diffusion Magnetic Resonance Imaging/methods , Disease Models, Animal , Glucose/metabolism , Magnetic Resonance Angiography/methods , Male , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods
15.
J Nucl Med ; 58(10): 1659-1665, 2017 10.
Article in English | MEDLINE | ID: mdl-28596158

ABSTRACT

Molecular imaging is indispensable for determining the fate and persistence of engrafted stem cells. Standard strategies for transgene induction involve the use of viral vectors prone to silencing and insertional mutagenesis or the use of nonhuman genes. Methods: We used zinc finger nucleases to induce stable expression of human imaging reporter genes into the safe-harbor locus adeno-associated virus integration site 1 in human embryonic stem cells. Plasmids were generated carrying reporter genes for fluorescence, bioluminescence imaging, and human PET reporter genes. Results: In vitro assays confirmed their functionality, and embryonic stem cells retained differentiation capacity. Teratoma formation assays were performed, and tumors were imaged over time with PET and bioluminescence imaging. Conclusion: This study demonstrates the application of genome editing for targeted integration of human imaging reporter genes in human embryonic stem cells for long-term molecular imaging.


Subject(s)
Embryonic Stem Cells/metabolism , Endoribonucleases/metabolism , Gene Editing , Genes, Reporter/genetics , Genome, Human/genetics , Positron-Emission Tomography , Zinc Fingers , Animals , Cell Differentiation , Cell Line , Endoribonucleases/chemistry , Female , Gene Expression , Humans , Liver/cytology , Mice
16.
Stem Cells Int ; 2016: 4095072, 2016.
Article in English | MEDLINE | ID: mdl-26880961

ABSTRACT

Tumor infiltrating stem cells have been suggested as a vehicle for the delivery of a suicide gene towards otherwise difficult to treat tumors like glioma. We have used herpes simplex virus thymidine kinase expressing human multipotent adult progenitor cells in two brain tumor models (hU87 and Hs683) in immune-compromised mice. In order to determine the best time point for the administration of the codrug ganciclovir, the stem cell distribution and viability were monitored in vivo using bioluminescence (BLI) and magnetic resonance imaging (MRI). Treatment was assessed by in vivo BLI and MRI of the tumors. We were able to show that suicide gene therapy using HSV-tk expressing stem cells can be followed in vivo by MRI and BLI. This has the advantage that (1) outliers can be detected earlier, (2) GCV treatment can be initiated based on stem cell distribution rather than on empirical time points, and (3) a more thorough follow-up can be provided prior to and after treatment of these animals. In contrast to rodent stem cell and tumor models, treatment success was limited in our model using human cell lines. This was most likely due to the lack of immune components in the immune-compromised rodents.

17.
Stem Cell Res Ther ; 6: 163, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26345383

ABSTRACT

INTRODUCTION: In this study, we planned to assess if adult stem cell-based suicide gene therapy can efficiently eliminate glioblastoma cells in vivo. We investigated the therapeutic potential of mouse Oct4(-) bone marrow multipotent adult progenitor cells (mOct4(-) BM-MAPCs) in a mouse glioblastoma model, guided by multimodal in vivo imaging methods to identify therapeutic windows. METHODS: Magnetic resonance imaging (MRI) of animals, wherein 5 × 10(5) syngeneic enhanced green fluorescent protein-firefly luciferase-herpes simplex virus thymidine kinase (eGFP-fLuc-HSV-TK) expressing and superparamagnetic iron oxide nanoparticle labeled (1 % or 10 %) mOct4(-) BM-MAPCs were grafted in glioblastoma (GL261)-bearing animals, showed that labeled mOct4(-) BM-MAPCs were located in and in close proximity to the tumor. Subsequently, ganciclovir (GCV) treatment was commenced and the fate of both the MAPCs and the tumor were followed by multimodal imaging (MRI and bioluminescence imaging). RESULTS: In the majority of GCV-treated, but not phosphate-buffered saline-treated animals, a significant difference was found in mOct4(-) BM-MAPC viability and tumor size at the end of treatment. Noteworthy, in some phosphate-buffered saline-treated animals (33 %), a significant decrease in tumor size was seen compared to sham-operated animals, which could potentially also be caused by a synergistic effect of the immune-modulatory stem cells. CONCLUSIONS: Suicide gene therapy using mOct4(-) BM-MAPCs as cellular carriers was effective in reducing the tumor size in the majority of the GCV-treated animals leading to a longer progression-free survival compared to sham-operated animals. This treatment could be followed and guided noninvasively in vivo by MRI and bioluminescence imaging. Noninvasive imaging is of particular interest for a rapid and efficient validation of stem cell-based therapeutic approaches for glioblastoma and hereby contributes to a better understanding and optimization of a promising therapeutic approach for glioblastoma patients.


Subject(s)
Bystander Effect , Genetic Therapy , Glioblastoma/therapy , Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/transplantation
18.
Stem Cells Dev ; 24(3): 296-311, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25203005

ABSTRACT

Cell-based therapies are emerging as an alternative treatment option to promote functional recovery in patients suffering from neurological disorders, which are the major cause of death and permanent disability. The present study aimed to differentiate human dental pulp stem cells (hDPSCs) toward functionally active neuronal cells in vitro. hDPSCs were subjected to a two-step protocol. First, neuronal induction was acquired through the formation of neurospheres, followed by neuronal maturation, based on cAMP and neurotrophin-3 (NT-3) signaling. At the ultrastructural level, it was shown that the intra-spheral microenvironment promoted intercellular communication. hDPSCs grew out of the neurospheres in vitro and established a neurogenic differentiated hDPSC culture (d-hDPSCs) upon cAMP and NT-3 signaling. d-hDPSCs were characterized by the increased expression of neuronal markers such as neuronal nuclei, microtubule-associated protein 2, neural cell adhesion molecule, growth-associated protein 43, synapsin I, and synaptophysin compared with nondifferentiated hDPSCs. Enzyme-linked immunosorbent assay demonstrated that the secretion of brain-derived neurotrophic factor, vascular endothelial growth factor, and nerve growth factor differed between d-hDPSCs and hDPSCs. d-hDPSCs acquired neuronal features, including multiple intercommunicating cytoplasmic extensions and increased vesicular transport, as shown by the electron microscopic observation. Patch clamp analysis demonstrated the functional activity of d-hDPSCs by the presence of tetrodotoxin- and tetraethyl ammonium-sensitive voltage-gated sodium and potassium channels, respectively. A subset of d-hDPSCs was able to fire a single action potential. The results reported in this study demonstrate that hDPSCs are capable of neuronal commitment following neurosphere formation, characterized by distinct morphological and electrophysiological properties of functional neuronal cells.


Subject(s)
Dental Pulp/cytology , Neurogenesis , Neurons/cytology , Action Potentials , Adolescent , Adult , Cell Communication , Cell Culture Techniques , Cell Separation/methods , Cell Shape , Cells, Cultured , Cellular Microenvironment , Culture Media/pharmacology , Female , Humans , Ion Channels/physiology , Male , Microscopy, Electron , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Patch-Clamp Techniques , Spheroids, Cellular , Young Adult
19.
Stem Cell Reports ; 5(5): 918-931, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26455413

ABSTRACT

Tools for rapid and efficient transgenesis in "safe harbor" loci in an isogenic context remain important to exploit the possibilities of human pluripotent stem cells (hPSCs). We created hPSC master cell lines suitable for FLPe recombinase-mediated cassette exchange (RMCE) in the AAVS1 locus that allow generation of transgenic lines within 15 days with 100% efficiency and without random integrations. Using RMCE, we successfully incorporated several transgenes useful for lineage identification, cell toxicity studies, and gene overexpression to study the hepatocyte lineage. However, we observed unexpected and variable transgene expression inhibition in vitro, due to DNA methylation and other unknown mechanisms, both in undifferentiated hESC and differentiating hepatocytes. Therefore, the AAVS1 locus cannot be considered a universally safe harbor locus for reliable transgene expression in vitro, and using it for transgenesis in hPSC will require careful assessment of the function of individual transgenes.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Targeting/methods , Hepatocytes/cytology , Induced Pluripotent Stem Cells/metabolism , Recombinases/metabolism , Transgenes , Cells, Cultured , DNA Methylation , Dependovirus/genetics , Embryonic Stem Cells/cytology , Gene Silencing , Genetic Loci , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Recombinases/genetics
20.
Pharmacol Ther ; 143(2): 181-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24594234

ABSTRACT

Mesenchymal stem cells or multipotent stromal cells (MSCs) have initially captured attention in the scientific world because of their differentiation potential into osteoblasts, chondroblasts and adipocytes and possible transdifferentiation into neurons, glial cells and endothelial cells. This broad plasticity was originally hypothesized as the key mechanism of their demonstrated efficacy in numerous animal models of disease as well as in clinical settings. However, there is accumulating evidence suggesting that the beneficial effects of MSCs are predominantly caused by the multitude of bioactive molecules secreted by these remarkable cells. Numerous angiogenic factors, growth factors and cytokines have been discovered in the MSC secretome, all have been demonstrated to alter endothelial cell behavior in vitro and induce angiogenesis in vivo. As a consequence, MSCs have been widely explored as a promising treatment strategy in disorders caused by insufficient angiogenesis such as chronic wounds, stroke and myocardial infarction. In this review, we will summarize into detail the angiogenic factors found in the MSC secretome and their therapeutic mode of action in pathologies caused by limited blood vessel formation. Also the application of MSC as a vehicle to deliver drugs and/or genes in (anti-)angiogenesis will be discussed. Furthermore, the literature describing MSC transdifferentiation into endothelial cells will be evaluated critically.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic/physiology , Animals , Cardiovascular Diseases/physiopathology , Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Cytokines/metabolism , Endothelial Cells/metabolism , Gene Transfer Techniques , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cell Transplantation , Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL