Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 16(26): 33246-33258, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38905518

ABSTRACT

Herein we report the assessment of the effects of shockwave (SW) impacts on adult rat hippocampal progenitor cell (AHPC) neurospheres (NSs), which are used as in vitro brain models, for enhancing our understanding of the mechanisms of traumatic brain injury (TBI). The assessment has been achieved by using culture dishes and a new microchip. The microchip allows the chemicals released from the brain models cultured inside the cell culture chamber under SW impacts to diffuse to the nanosensors in adjacent sensor chambers through built-in diffusion barriers, which are used to prevent the cells from entering the sensor chambers, thereby mitigating the biofouling issues of the sensor surface. Experiments showed the negative impact of the SW on the viability, proliferation, and differentiation of the cells within the NSs. A qPCR gene expression analysis was performed and appeared to confirm some of the immunocytochemistry (ICC) results. Finally, we demonstrated that the microchip can be used to monitor lactate dehydrogenase (LDH) released from the AHPC-NSs subjected to SW impacts. As expected, LDH levels changed when AHPC-NSs were injured by SW impacts, verifying this chip can be used for assessing the degrees of injuries to AHPC-NSs by monitoring LDH levels. Taken together, these results suggest the feasibility of using the chip to better understand the interactions between SW impacts and in vitro brain models, paving the way for potentially establishing in vitro TBI models on a chip.


Subject(s)
Brain Injuries, Traumatic , Hippocampus , Animals , Rats , Hippocampus/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Lab-On-A-Chip Devices , Cell Survival , L-Lactate Dehydrogenase/metabolism , Cell Proliferation , Brain/metabolism , Brain/pathology , High-Energy Shock Waves , Cells, Cultured , Cell Differentiation
2.
Lab Chip ; 23(6): 1649-1663, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36751868

ABSTRACT

To improve our understanding of how the central nervous system functions in health and disease, we report the development of an integrated chip for studying the effects of the neurotransmitters dopamine and serotonin on adult rat hippocampal progenitor cell (AHPC) neurospheroids. This chip allows dopamine or serotonin located in one chamber to diffuse to AHPC neurospheroids cultured in an adjacent chamber through a built-in diffusion barrier created by an array of intentionally misaligned micropillars. The gaps among the micropillars are filled with porous poly(ethylene glycol) (PEG) gel to tune the permeability of the diffusion barrier. An electrochemical sensor is also integrated within the chamber where the neurospheroids can be cultured, thereby allowing monitoring of the concentrations of dopamine or serotonin. Experiments show that concentrations of the neurotransmitters inside the neurospheroid chamber can be increased over a period of several hours to over 10 days by controlling the compositions of the PEG gel inside the diffusion barrier. The AHPC neurospheroids cultured in the chip remain highly viable following dopamine or serotonin treatment. Cell proliferation and neuronal differentiation have also been observed following treatment, revealing that the AHPC neurospheroids are a valuable in vitro brain model for neurogenesis research. Finally, we show that by tuning the permeability of diffusion barrier, we can block transfer of Escherichia coli cells across the diffusion barrier, while allowing dopamine or serotonin to pass through. These results suggest the feasibility of using the chip to better understand the interactions between microbiota and brain via the gut-brain axis.


Subject(s)
Dopamine , Microfluidics , Rats , Animals , Serotonin , Brain , Neurotransmitter Agents
3.
Am J Pathol ; 190(7): 1461-1473, 2020 07.
Article in English | MEDLINE | ID: mdl-32259521

ABSTRACT

Proteinopathies result from aberrant folding and accumulation of specific proteins. Currently, there is a lack of knowledge about the factors that influence disease progression, making this a key challenge for the development of therapies for proteinopathies. Because of the similarities between transmissible spongiform encephalopathies (TSEs) and other protein misfolding diseases, TSEs can be used to understand other proteinopathies. Bovine spongiform encephalopathy (BSE) is a TSE that occurs in cattle and can be subdivided into three strains: classic BSE and atypical BSEs (H and L types) that have shorter incubation periods. The NACHT, LRR, and PYD domains-containing protein 3 inflammasome is a critical component of the innate immune system that leads to release of IL-1ß. Macroautophagy is an intracellular mechanism that plays an essential role in protein clearance. In this study, the retina was used as a model to investigate the relationship between disease incubation period, prion protein accumulation, neuroinflammation, and changes in macroautophagy. We demonstrate that atypical BSEs present with increased prion protein accumulation, neuroinflammation, and decreased autophagy. This work suggests a relationship between disease time course, neuroinflammation, and the autophagic stress response, and may help identify novel therapeutic biomarkers that can delay or prevent the progression of proteinopathies.


Subject(s)
Autophagy/physiology , Encephalopathy, Bovine Spongiform/pathology , Inflammation/pathology , PrPSc Proteins/pathogenicity , Animals , Cattle , Encephalopathy, Bovine Spongiform/immunology , Inflammation/immunology , Male , Proteostasis Deficiencies/immunology , Proteostasis Deficiencies/pathology , Retina/immunology , Retina/pathology
4.
ACS Omega ; 5(14): 7910-7918, 2020 Apr 14.
Article in English | MEDLINE | ID: mdl-32309700

ABSTRACT

Because of the limitations imposed by traditional two-dimensional (2D) cultures, biomaterials have become a major focus in neural and tissue engineering to study cell behavior in vitro. 2D systems fail to account for interactions between cells and the surrounding environment; these cell-matrix interactions are important to guide cell differentiation and influence cell behavior such as adhesion and migration. Biomaterials provide a unique approach to help mimic the native microenvironment in vivo. In this study, a novel microfluidic technique is used to encapsulate adult rat hippocampal stem/progenitor cells (AHPCs) within alginate-based fibrous hydrogels. To our knowledge, this is the first study to encapsulate AHPCs within a fibrous hydrogel. Alginate-based hydrogels were cultured for 4 days in vitro and recovered to investigate the effects of a 3D environment on the stem cell fate. Post recovery, cells were cultured for an additional 24 or 72 h in vitro before fixing cells to determine if proliferation and neuronal differentiation were impacted after encapsulation. The results indicate that the 3D environment created within a hydrogel is one factor promoting AHPC proliferation and neuronal differentiation (19.1 and 13.5%, respectively); however, this effect is acute. By 72 h post recovery, cells had similar levels of proliferation and neuronal differentiation (10.3 and 8.3%, respectively) compared to the control conditions. Fibrous hydrogels may better mimic the natural micro-environment present in vivo and be used to encapsulate AHPCs, enhancing cell proliferation and selective differentiation. Understanding cell behavior within 3D scaffolds may lead to the development of directed therapies for central nervous system repair and rescue.

5.
J Neurosci Methods ; 328: 108419, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31472190

ABSTRACT

BACKGROUND: Primary cell culture is a valuable tool to utilize in parallel with in vivo studies in order to maximize our understanding of the mechanisms surrounding neurogenesis and central nervous system (CNS) regeneration and plasticity. The zebrafish is an important model for biomedical research and primary neural cells are readily obtainable from their embryonic stages viatissue dissociation. Further, transgenic reporter lines with cell type-specific expression allows for observation of distinct cell populations within the dissociated tissue. NEW METHOD: Here, we define an efficient method for ex vivo quantification and characterization of neuronal and glial tissue dissociated from embryonic zebrafish. RESULTS: Zebrafish brain dissociated cells have been documented to survive in culture for at least 9 days in vitro (div). Anti-HuC/D and anti-Acetylated Tubulin antibodies were used to identify neurons in culture; at 3 div approximately 48% of cells were HuC/D positive and 85% expressed serotonin, suggesting our protocol can efficiently isolate neurons from whole embryonic zebrafish brains. Live time-lapse imaging was also carried out to analyze cell migration in vitro. COMPARISON WITH EXISTING METHODS: Primary cultures of zebrafish neural cells typically have low rates of survivability in vitro. We have developed a culture system that has long term cell viability, enabling direct analysis of cell-cell and cell-extracellular matrix interactions. CONCLUSIONS: These results demonstrate a practical method for isolating, dissociating and culturing of embryonic zebrafish neural tissue. This approach could further be utilized to better understand zebrafish regeneration in vitro.


Subject(s)
Neuroglia , Neurons , Neurosciences/methods , Primary Cell Culture/methods , Zebrafish , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Neuroglia/cytology , Neuroglia/physiology , Neurons/cytology , Neurons/physiology
6.
ACS Appl Mater Interfaces ; 11(20): 18797-18807, 2019 May 22.
Article in English | MEDLINE | ID: mdl-31042026

ABSTRACT

Bow tie-shaped fibers and spherical microparticles with controlled dimensions and shapes were fabricated with poly(ethylene glycol) diacrylate hydrogel utilizing hydrodynamic shear principles and a photopolymerization strategy under a microfluidic regime. Decreasing the flow rate ratio between the core and sheath fluids from 25 (50:2) to 1.25 (100:80) resulted in increasing the particles size and reducing the production rate by 357 and 86%, respectively. The width of the fibers increased by a factor of 1.4 when the flow rate ratio was reduced from 2.5 to 1 due to the decrease of the shear force at the fluid/fluid interface. The stress at break and Young's modulus of the fibers were enhanced by 32 and 63%, respectively, when the sheath-to-core flow rate ratio decreased from 100:40 to 100:80. The fiber fabrication was simulated using the finite element method, and the numerical and experimental results were in agreement. Adult hippocampal stem/progenitor cells and bone-marrow-derived multipotent mesenchymal stromal cells were seeded onto the fibrous scaffolds in vitro, and cellular adhesion, proliferation, and differentiation were investigated. Microgrooves on the fibers' surface were shown to positively affect cell adhesion when compared to flat fibers and planar controls.

7.
Methods Mol Biol ; 1940: 143-155, 2019.
Article in English | MEDLINE | ID: mdl-30788823

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs) hold great potential as an ex vivo cellular system for delivery of therapeutic proteins to the diseased or damaged central nervous system (CNS). This adult stem cell population has considerable translational potential for autologous transplantation due to lack of ethical concerns, accessibility, multipotent nature, and plasticity. Here we describe a methodology and outline a strategy using lentiviral vectors for producing lines of MSCs hypersecreting neurotrophic growth factors (e.g., brain-derived neurotrophic factor (BDNF) and/or glial cell line-derived neurotrophic factor (GDNF)) together with a reporter protein such as green fluorescent protein (GFP) that may be used for in vitro and in vivo neuroprotection bioassays. This approach provides exciting opportunities for basic research and proof-of-concept studies.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell- and Tissue-Based Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Central Nervous System/pathology , Genetic Engineering/methods , Glial Cell Line-Derived Neurotrophic Factor/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lentivirus/genetics , Mice , Neurodegenerative Diseases/therapy , Retina/transplantation
8.
Macromol Biosci ; 19(2): e1800236, 2019 02.
Article in English | MEDLINE | ID: mdl-30480879

ABSTRACT

Biomaterials are essential for the development of innovative biomedical and therapeutic applications. Biomaterials-based scaffolds can influence directed cell differentiation to improve cell-based strategies. Using a novel microfluidics approach, poly (ε-caprolactone) (PCL), is used to fabricate microfibers with varying diameters (3-40 µm) and topographies (straight and wavy). Multipotent adult rat hippocampal stem/progenitor cells (AHPCs) are cultured on 3D aligned PCL microfibrous scaffolds to investigate their ability to differentiate into neurons, astrocytes, and oligodendrocytes. The results indicate that the PCL microfibers significantly enhance proliferation of the AHPCs compared to control, 2D planar substrates. While the AHPCs maintained their multipotent differentiation capacity when cultured on the PCL scaffolds, there is a significant and dramatic increase in immunolabeling for astrocyte and oligodendrocyte differentiation when compared with growth on planar surfaces. Our results show a 3.5-fold increase in proliferation and 23.4-fold increase in astrocyte differentiation for cells on microfibers. Transplantation of neural stem/progenitor cells within a PCL microfiber scaffold may provide important biological and topographic cues that facilitate the survival, selective differentiation, and integration of transplanted cells to improve therapeutic strategies.


Subject(s)
Adult Stem Cells/cytology , Astrocytes/cytology , Neural Stem Cells/cytology , Neurons/cytology , Oligodendroglia/cytology , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Brain Injuries/therapy , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Hippocampus/cytology , Methacrylates/chemistry , Microfluidics/methods , Neurodegenerative Diseases/therapy , Neurogenesis/physiology , Polyesters/chemistry , Rats , Tissue Scaffolds/chemistry
9.
Neurobiol Dis ; 121: 1-16, 2019 01.
Article in English | MEDLINE | ID: mdl-30218757

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by accumulation of misfolded α-synuclein within the central nervous system (CNS). Visual problems in PD patients are common, although retinal pathology associated with PD is not well understood. The purpose of this study was to investigate retinal pathology in a transgenic mouse model (TgM83) expressing the human A53T α-synuclein mutation and assess the effect of α-synuclein "seeding" on the development of retinal pathology. Two-month-old TgM83 mice were intracerebrally inoculated with brain homogenate from old (12-18 months) TgM83 mice. Retinas were then analyzed at 5 months of age. We analyzed retinas from 5-month-old and 8-month-old uninoculated healthy TgM83 mice, and old (12-18 months) mice that were euthanized following the development of clinical signs. Retinas of B6C3H mice (genetic background of the TgM83 mouse) served as control. We used immunohistochemistry and western blot analysis to detect accumulation of α-synuclein, pTauThr231, inflammation, changes in macroautophagy, and cell death. Raman spectroscopy was used to test the potential to differentiate between retinal tissues of healthy mice and diseased mice. This work demonstrates retinal changes associated with the A53T mutation. Retinas of non-inoculated TgM83 mice had accumulation of α-synuclein, "pre-tangle" tau, activation of retinal glial cells, and photoreceptor cell loss by 8 months of age. The development of these changes is accelerated by inoculation with brain homogenate from clinically ill TgM83 mice. Compared to non-inoculated 5-month-old TgM83 mice, retinas of inoculated 5-month-old mice had increased accumulation of α-synuclein (pSer129) and pTauThr231 proteins, upregulated microglial activation, and dysregulated macroautophagy. Raman spectroscopic analysis was able to discriminate between healthy and diseased mice. This study describes retinal pathology resulting from the A53T mutation. We show that seeding with brain homogenates from old TgM83 mice accelerates retinal pathology. We demonstrate that Raman spectroscopy can be used to accurately identify a diseased retina based on its biochemical profile, and that α-synuclein accumulation may contribute to accumulation of pTauThr231 proteins, neuroinflammation, metabolic dysregulation, and photoreceptor cell death. Our work provides insight into retinal changes associated with Parkinson's disease, and may contribute to a better understanding of visual symptoms experienced by patients.


Subject(s)
Autophagy , Encephalitis/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Retina/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Encephalitis/complications , Mice, Transgenic , Neuroglia/metabolism , Parkinson Disease/complications , Phosphorylation , Retina/pathology
10.
Neural Regen Res ; 14(2): 256-257, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30531005
11.
Adv Exp Med Biol ; 1119: 41-71, 2018.
Article in English | MEDLINE | ID: mdl-30151648

ABSTRACT

Peripheral nerve injuries (PNI) occur as the result of sudden trauma and can lead to life-long disability, reduced quality of life, and heavy economic and social burdens. Although the peripheral nervous system (PNS) has the intrinsic capacity to regenerate and regrow axons to a certain extent, current treatments frequently show incomplete recovery with poor functional outcomes, particularly for large PNI. Many surgical procedures are available to halt the propagation of nerve damage, and the choice of a procedure depends on the extent of the injury. In particular, recovery from large PNI gaps is difficult to achieve without any therapeutic intervention or some form of tissue/cell-based therapy. Autologous nerve grafting, considered the "gold standard" is often implemented for treatment of gap formation type PNI. Although these surgical procedures provide many benefits, there are still considerable limitations associated with such procedures as donor site morbidity, neuroma formation, fascicle mismatch, and scarring. To overcome such restrictions, researchers have explored various avenues to improve post-surgical outcomes. The most commonly studied methods include: cell transplantation, growth factor delivery to stimulate regenerating axons and implanting nerve guidance conduits containing replacement cells at the site of injury. Replacement cells which offer maximum benefits for the treatment of PNI, are Schwann cells (SCs), which are the peripheral glial cells and in part responsible for clearing out debris from the site of injury. Additionally, they release growth factors to stimulate myelination and axonal regeneration. Both primary SCs and genetically modified SCs enhance nerve regeneration in animal models; however, there is no good source for extracting SCs and the only method to obtain SCs is by sacrificing a healthy nerve. To overcome such challenges, various cell types have been investigated and reported to enhance nerve regeneration.In this review, we have focused on cell-based strategies aimed to enhance peripheral nerve regeneration, in particular the use of mesenchymal stem cells (MSCs). Mesenchymal stem cells are preferred due to benefits such as autologous transplantation, routine isolation procedures, and paracrine and immunomodulatory properties. Mesenchymal stem cells have been transplanted at the site of injury either directly in their native form (undifferentiated) or in a SC-like form (transdifferentiated) and have been shown to significantly enhance nerve regeneration. In addition to transdifferentiated MSCs, some studies have also transplanted ex-vivo genetically modified MSCs that hypersecrete growth factors to improve neuroregeneration.


Subject(s)
Adult Stem Cells , Peripheral Nerve Injuries , Animals , Nerve Regeneration , Peripheral Nerves , Quality of Life , Schwann Cells
12.
PLoS One ; 13(5): e0198025, 2018.
Article in English | MEDLINE | ID: mdl-29795671

ABSTRACT

Adult stem cells are considered multipotent, restricted to differentiate into a few tissue-specific cell types. With the advent of technologies which can dedifferentiate and transdifferentiate cell types, assumptions about the process of cell fate determination must be reconsidered, including the role of extrinsic versus intrinsic factors. To determine the plasticity of adult neural progenitors, rat hippocampal progenitor cells were xenotransplanted into embryonic zebrafish. These animals allow for easy detection of transplanted cells due to their external development and transparency at early stages. Adult neural progenitors were observed throughout the zebrafish for the duration of the experiment (at least five days post-transplantation). While the majority of transplanted cells were observed in the central nervous system, a large percentage of cells were located in superficial tissues. However, approximately one-third of these cells retained neural morphology and expression of the neuronal marker, Class III ß-tubulin, indicating that the transplanted adult neural progenitors did not adapt alternate fates. A very small subset of cells demonstrated unique, non-neural flattened morphology, suggesting that adult neural progenitors may exhibit plasticity in this model, though at a very low rate. These findings demonstrate that the developing zebrafish may be an efficient model to explore plasticity of a variety of adult stem cell types and the role of external factors on cell fate.


Subject(s)
Cell Differentiation , Cell Plasticity , Embryo, Nonmammalian/cytology , Hippocampus/cytology , Neural Stem Cells/cytology , Zebrafish/embryology , Animals , Cells, Cultured , Embryo, Nonmammalian/physiology , Hippocampus/physiology , Neural Stem Cells/physiology , Rats , Transplantation, Heterologous
13.
Adv Healthc Mater ; 7(14): e1701046, 2018 07.
Article in English | MEDLINE | ID: mdl-29656561

ABSTRACT

Adult stems cells, possessing the ability to grow, migrate, proliferate, and transdifferentiate into various specific phenotypes, constitute a great asset for peripheral nerve regeneration. Adult stem cells' ability to undergo transdifferentiation is sensitive to various cell-to-cell interactions and external stimuli involving interactions with physical, mechanical, and chemical cues within their microenvironment. Various studies have employed different techniques for transdifferentiating adult stem cells from distinct sources into specific lineages (e.g., glial cells and neurons). These techniques include chemical and/or electrical induction as well as cell-to-cell interactions via co-culture along with the use of various 3D conduit/scaffold designs. Such scaffolds consist of unique materials that possess controllable physical/mechanical properties mimicking cells' natural extracellular matrix. However, current limitations regarding non-scalable transdifferentiation protocols, fate commitment of transdifferentiated stem cells, and conduit/scaffold design have required new strategies for effective stem cells transdifferentiation and implantation. In this progress report, a comprehensive review of recent advances in the transdifferentiation of adult stem cells via different approaches along with multifunctional conduit/scaffolds designs is presented for peripheral nerve regeneration. Potential cellular mechanisms and signaling pathways associated with differentiation are also included. The discussion with current challenges in the field and an outlook toward future research directions is concluded.


Subject(s)
Adult Stem Cells/cytology , Nerve Regeneration/physiology , Peripheral Nerves/physiology , Adult Stem Cells/physiology , Animals , Biocompatible Materials/chemistry , Cell Communication/physiology , Cell Differentiation/physiology , Humans , Tissue Scaffolds/chemistry
14.
J Biosci Bioeng ; 124(5): 572-582, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28694020

ABSTRACT

The use of genetically modified mesenchymal stem cells (MSCs) is a rapidly growing area of research targeting delivery of therapeutic factors for neuro-repair. Cells can be programmed to hypersecrete various growth/trophic factors such as brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and nerve growth factor (NGF) to promote regenerative neurite outgrowth. In addition to genetic modifications, MSCs can be subjected to transdifferentiation protocols to generate neural cell types to physically and biologically support nerve regeneration. In this study, we have taken a novel approach by combining these two unique strategies and evaluated the impact of transdifferentiating genetically modified MSCs into a Schwann cell-like phenotype. After 8 days in transdifferentiation media, approximately 30-50% of transdifferentiated BDNF-secreting cells immunolabeled for Schwann cell markers such as S100ß, S100, and p75NTR. An enhancement was observed 20 days after inducing transdifferentiation with minimal decreases in expression levels. BDNF production was quantified by ELISA, and its biological activity tested via the PC12-TrkB cell assay. Importantly, the bioactivity of secreted BDNF was verified by the increased neurite outgrowth of PC12-TrkB cells. These findings demonstrate that not only is BDNF actively secreted by the transdifferentiated BDNF-MSCs, but also that it has the capacity to promote neurite sprouting and regeneration. Given the fact that BDNF production remained stable for over 20 days, we believe that these cells have the capacity to produce sustainable, effective, BDNF concentrations over prolonged time periods and should be tested within an in vivo system for future experiments.


Subject(s)
Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Transdifferentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Schwann Cells/cytology , Schwann Cells/metabolism , Animals , Cell Transdifferentiation/drug effects , Culture Media/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mice , Nerve Regeneration , Neurites/drug effects , Neurites/physiology , PC12 Cells , Rats , Receptor, trkB/metabolism , Schwann Cells/drug effects
15.
J Proteomics ; 165: 93-101, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28629798

ABSTRACT

While transplantation of Schwann cells facilitates axon regeneration, remyelination and repair after peripheral nerve injury clinical use is limited by cell bioavailability. We posit that such limitation in cell access can be overcome by the use of autologous bone-marrow derived mesenchymal stem cells (MSCs). As MSCs can transdifferentiate to Schwann cell-phenotypes and accelerate nerve regeneration we undertook proteomic evaluation of the cells to uncover the protein contents that affects Schwann cell formulation. Transdifferentiated MSCs secrete significant amounts of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in cell-conditioned media that facilitated neurite outgrowth. MSC proteins significantly regulated during Schwann cell transdifferentiation included, but were not limited to, GNAI2, MYL9, ACTN4, ACTN1, ACTB, CAV-1, HSPB1, PHB2, TBB4B, CTGF, TGFI1, ARF6, EZR, GELS, VIM, WNT5A, RTN4, EFNB1. These support axonal guidance, myelination, neural development and neural growth and differentiation. The results unravel the molecular events that underlie cell transdifferentiation that ultimately serve to facilitate nerve regeneration and repair in support of cell transplantation. SIGNIFICANCE STATEMENT: While Schwann cells facilitate axon regeneration, remyelination and repair after peripheral nerve injury clinical use is limited by cell bioavailability. We posit that such limitation in cell access can be overcome by the use of bone-marrow derived mesenchymal stem cells (MSCs) transdifferentiated to Schwann cell-phenotypes. In the present study, we undertook the first proteomic evaluation of these transdifferentiated cells to uncover the protein contents that affects Schwann cell formulation. Furthermore, these transdifferentiated MSCs secrete significant amounts of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in cell-conditioned media that facilitated neurite outgrowth. Our results demonstrate that a number of MSC proteins were significantly regulated following transdifferentiation of the MSCs supporting roles in axonal guidance, myelination, neural development and differentiation. The conclusions of the present work unravel the molecular events that underlie cell transdifferentiation that ultimately serve to facilitate nerve regeneration and repair in support of cell transplantation. Our study was the first proteomic comparison demonstrating the transdifferentiation of MSCs and these reported results can affect a wide field of stem cell biology, tissue engineering, and proteomics.


Subject(s)
Cell Transdifferentiation , Mesenchymal Stem Cells/cytology , Proteomics/methods , Schwann Cells/cytology , Animals , Brain-Derived Neurotrophic Factor/analysis , Cells, Cultured , Mesenchymal Stem Cells/chemistry , Nerve Growth Factor/analysis , Nerve Regeneration , Rats , Schwann Cells/chemistry
16.
Am J Pathol ; 187(7): 1459-1472, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28606756

ABSTRACT

Traumatic brain injury due to blast exposure is currently the most prevalent of war injuries. Although secondary ocular blast injuries due to flying debris are more common, primary ocular blast exposure resulting from blast wave pressure has been reported among survivors of explosions, but with limited understanding of the resulting retinal pathologies. Using a compressed air-driven shock tube system, adult male and female C57BL/6 mice were exposed to blast wave pressure of 300 kPa (43.5 psi) per day for 3 successive days, and euthanized 30 days after injury. We assessed retinal tissues using immunofluorescence for glial fibrillary acidic protein, microglia-specific proteins Iba1 and CD68, and phosphorylated tau (AT-270 pThr181 and AT-180 pThr231). Primary blast wave pressure resulted in activation of Müller glia, loss of photoreceptor cells, and an increase in phosphorylated tau in retinal neurons and glia. We found that 300-kPa blasts yielded no detectable cognitive or motor deficits, and no neurochemical or biochemical evidence of injury in the striatum or prefrontal cortex, respectively. These changes were detected 30 days after blast exposure, suggesting the possibility of long-lasting retinal injury and neuronal inflammation after primary blast exposure.


Subject(s)
Blast Injuries/physiopathology , Calcium-Binding Proteins/metabolism , High-Energy Shock Waves/adverse effects , Microfilament Proteins/metabolism , Retinal Diseases/physiopathology , Wounds and Injuries/physiopathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blast Injuries/metabolism , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Retina/injuries , Retinal Diseases/metabolism , Time Factors , Wounds and Injuries/metabolism , tau Proteins/metabolism
17.
Zebrafish ; 14(4): 343-356, 2017 08.
Article in English | MEDLINE | ID: mdl-28192065

ABSTRACT

In the central nervous system injury induces cellular reprogramming and progenitor proliferation, but the molecular mechanisms that limit regeneration and prevent tumorigenesis are not completely understood. We previously described a zebrafish optic pathway tumor model in which transgenic Tg(flk1:RFP)is18/+ adults develop nonmalignant retinal tumors. Key pathways driving injury-induced glial reprogramming and regeneration contributed to tumor formation. In this study, we examine a time course of proliferation and present new analyses of the Tg(flk1:RFP)is18/+ dysplastic retina and tumor transcriptomes. Retinal dysplasia was first detected in 3-month-old adults, but was not limited to a specific stem cell or progenitor niche. Pathway analyses suggested a decrease in cellular respiration and increased expression of components of Hif1-α, VEGF, mTOR, NFκß, and multiple interleukin pathways are associated with early retinal dysplasia. Hif-α targets VEGFA (vegfab) and Leptin (lepb) were both highly upregulated in dysplastic retina; however, each showed distinct expression patterns in neurons and glia, respectively. Phospho-S6 immunolabeling indicated that mTOR signaling is activated in multiple cell populations in wild-type retina and in the dysplastic retina and advanced tumor. Our results suggest that multiple pathways may contribute to the continuous proliferation of retinal progenitors and tumor growth in this optic pathway tumor model. Further investigation of these signaling pathways may yield insight into potential mechanisms to control the proliferative response during regeneration in the nervous system.


Subject(s)
Cell Proliferation , Eye Neoplasms/pathology , Leptin/metabolism , Retinal Dysplasia/pathology , Vascular Endothelial Growth Factor A/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Eye Neoplasms/genetics , Eye Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Leptin/genetics , Retinal Dysplasia/genetics , Retinal Dysplasia/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
18.
Acta Biomater ; 53: 293-306, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28213098

ABSTRACT

In this study, gelatin-based 3D conduits with three different microstructures (nanofibrous, macroporous and ladder-like) were fabricated for the first time via combined molding and thermally induced phase separation (TIPS) technique for peripheral nerve regeneration. The effects of conduit microstructure and mechanical properties on the transdifferentiation of bone marrow-derived mesenchymal stem cells (MSCs) into Schwann cell (SC) like phenotypes were examined to help facilitate neuroregeneration and understand material-cell interfaces. Results indicated that 3D macroporous and ladder-like structures enhanced MSC attachment, proliferation and spreading, creating interconnected cellular networks with large numbers of viable cells compared to nanofibrous and 2D-tissue culture plate counterparts. 3D-ladder-like conduit structure with complex modulus of ∼0.4×106Pa and pore size of ∼150µm provided the most favorable microenvironment for MSC transdifferentiation leading to ∼85% immunolabeling of all SC markers. On the other hand, the macroporous conduits with complex modulus of ∼4×106Pa and pore size of ∼100µm showed slightly lower (∼65% for p75, ∼75% for S100 and ∼85% for S100ß markers) immunolabeling. Transdifferentiated MSCs within 3D-ladder-like conduits secreted significant amounts (∼2.5pg/mL NGF and ∼0.7pg/mL GDNF per cell) of neurotrophic factors, while MSCs in macroporous conduits released slightly lower (∼1.5pg/mL NGF and 0.7pg/mL GDNF per cell) levels. PC12 cells displayed enhanced neurite outgrowth in media conditioned by conduits with transdifferentiated MSCs. Overall, conduits with macroporous and ladder-like 3D structures are promising platforms in transdifferentiation of MSCs for neuroregeneration and should be further tested in vivo. STATEMENT OF SIGNIFICANCE: This manuscript focuses on the effect of microstructure and mechanical properties of gelatin-based 3D conduits on the transdifferentiation of mesenchymal stem cells to Schwann cell-like phenotypes. This work builds on our recently accepted manuscript in Acta Biomaterialia focused on multifunctional 2D films, and focuses on 3D microstructured conduits designed to overcome limitations of current strategies to facilitate peripheral nerve regeneration. The comparison between conduits fabricated with nanofibrous, macroporous and ladder-like microstructures showed that the ladder-like conduits showed the most favorable environment for MSC transdifferentiation to Schwann-cell like phenotypes, as seen by both immunolabeling as well as secretion of neurotrophic factors. This work demonstrates the importance of controlling the 3D microstructure to facilitate tissue engineering strategies involving stem cells that can serve as promising approaches for peripheral nerve regeneration.


Subject(s)
Cell Differentiation/physiology , Gelatin/chemistry , Guided Tissue Regeneration/instrumentation , Mesenchymal Stem Cell Transplantation/instrumentation , Mesenchymal Stem Cells/cytology , Printing, Three-Dimensional , Schwann Cells/cytology , Tissue Scaffolds , Animals , Cell Transdifferentiation/physiology , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Nerve Regeneration/physiology , Rats , Schwann Cells/physiology , Tissue Engineering/instrumentation , Tissue Engineering/methods
19.
Adv Healthc Mater ; 6(7)2017 Apr.
Article in English | MEDLINE | ID: mdl-28218474

ABSTRACT

Graphene-based materials (GBMs) have displayed tremendous promise for use as neurointerfacial substrates as they enable favorable adhesion, growth, proliferation, spreading, and migration of immobilized cells. This study reports the first case of the differentiation of mesenchymal stem cells (MSCs) into Schwann cell (SC)-like phenotypes through the application of electrical stimuli from a graphene-based electrode. Electrical differentiation of MSCs into SC-like phenotypes is carried out on a flexible, inkjet-printed graphene interdigitated electrode (IDE) circuit that is made highly conductive (sheet resistance < 1 kΩ/sq) via a postprint pulse-laser annealing process. MSCs immobilized on the graphene printed IDEs and electrically stimulated/treated (etMSCs) display significant enhanced cellular differentiation and paracrine activity above conventional chemical treatment strategies [≈85% of the etMSCs differentiated into SC-like phenotypes with ≈80 ng mL-1 of nerve growth factor (NGF) secretion vs. 75% and ≈55 ng mL-1 for chemically treated MSCs (ctMSCs)]. These results help pave the way for in vivo peripheral nerve regeneration where the flexible graphene electrodes could conform to the injury site and provide intimate electrical simulation for nerve cell regrowth.


Subject(s)
Cell Differentiation , Graphite/chemistry , Mesenchymal Stem Cells/metabolism , Schwann Cells/metabolism , Animals , Electric Stimulation , Mesenchymal Stem Cells/cytology , Rats , Schwann Cells/cytology
20.
Acta Biomater ; 56: 141-152, 2017 07 01.
Article in English | MEDLINE | ID: mdl-27693689

ABSTRACT

In this study, a poly(lactic acid) (PLLA) porous film with longitudinal surface micropatterns was fabricated by a dry phase inversion technique to be used as potential conduit material for peripheral nerve regeneration applications. The presence of a nerve growth factor (NGF) gradient on the patterned film surface and protein loaded, surface-eroding, biodegradable, and amphiphilic polyanhydride (PA) microparticles within the film matrix, enabled co-delivery of neurotrophic factors with controlled release properties and enhanced neurite outgrowth from PC12 cells. The protein loading capacity of PA particles was increased up to 80% using the spray drying technique, while the surface loading of NGF reached 300ng/cm2 through ester-amine interactions. The NGF surface gradient provided initial fast release from the film surface and facilitated directional neurite outgrowth along with the longitudinal micropatterns. Furthermore, the variable backbone chemistry and surface eroding nature of protein-loaded PA microparticles within the film matrix ensured protein stability and enabled controlled protein release. This novel co-delivery strategy yielded tunable diffusion coefficients varying between 6×10-14 and 1.67×10-10cm2/min and dissolution constants ranging from 1×10-4 to 1×10-3min-1 with released amounts of ∼100-300ng/mL. This strategy promoted guided neurite extension from PC12 cells of up to 10µm total neurite length per cell in 2days. Overall, this unique strategy can potentially be extended for individually programmed delivery of multiple growth factors through the use of PA microparticle cocktails and can further be investigated for in vivo performance as potential conduit material for peripheral nerve regeneration applications. STATEMENT OF SIGNIFICANCE: This manuscript focuses on the development of multifunctional degradable polymer films that provide topographic cues for guided growth, surface gradients of growth factors as well as nanoparticles in the films for tunable release of growth factors to enable peripheral nerve regeneration. The combination of cues was designed to overcome limitations of current strategies to facilitate peripheral nerve regeneration. These multifunctional films successfully provided high protein loading capacities while persevering activity, protein gradients on the surface, and tunable release of bioactive nerve growth factor that promoted directional and guided neurite extension of PC12 cells of up to 10µm in 2days. These multifunctional films can be made into conduits for peripheral nerve regeneration.


Subject(s)
Membranes, Artificial , Nerve Growth Factor , Nerve Regeneration/drug effects , Neurites/metabolism , Peripheral Nerves/physiology , Animals , Biodegradable Plastics/chemistry , Nerve Growth Factor/pharmacology , PC12 Cells , Polyesters/chemistry , Porosity , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...