Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 14(1): 83, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37046329

ABSTRACT

Stem cell replacement holds the potential for sensorineural hearing loss (SNHL) treatment. However, its translation into clinical practice requires strategies for improving stem cell survival following intracochlear transplantation. Considering recent findings showing that the inner ear contains a resident population of immune cells, we hypothesized that immune evasion would improve the survival and residence time of transplanted stem cells in the cochlea, potentially leading to better outcomes. To test this, we leveraged genetic engineering techniques to develop hypoimmunogenic human-induced pluripotent stem cells (hi-iPSC), which lack human leukocyte antigen expression. We found that gene editing does not affect the biological properties of hi-iPSCs, including their capacity to differentiate into otic neural progenitors (ONPs). Compared to wild-type ONPs, more hypoimmunogenic ONPs (derived from hi-iPSCs) were found in the inner ear of immunocompetent mice ten days following cochlear xenotransplantation. This approach may open a new avenue for experimental and clinical SNHL treatments.


Subject(s)
Hearing Loss , Induced Pluripotent Stem Cells , Mice , Humans , Animals , Transplantation, Heterologous , Cell Differentiation , Hearing Loss/metabolism , Stem Cell Transplantation/methods , Induced Pluripotent Stem Cells/metabolism
2.
Nat Commun ; 13(1): 2650, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551462

ABSTRACT

Ketamine treatment decreases depressive symptoms within hours, but the mechanisms mediating these rapid antidepressant effects are unclear. Here, we demonstrate that activity of adult-born immature granule neurons (ABINs) in the mouse hippocampal dentate gyrus is both necessary and sufficient for the rapid antidepressant effects of ketamine. Ketamine treatment activates ABINs in parallel with its behavioral effects in both stressed and unstressed mice. Chemogenetic inhibition of ABIN activity blocks the antidepressant effects of ketamine, indicating that this activity is necessary for the behavioral effects. Conversely, chemogenetic activation of ABINs without any change in neuron numbers mimics both the cellular and the behavioral effects of ketamine, indicating that increased activity of ABINs is sufficient for rapid antidepressant effects. These findings thus identify a specific cell population that mediates the antidepressant actions of ketamine, indicating that ABINs can potentially be targeted to limit ketamine's side effects while preserving its therapeutic efficacy.


Subject(s)
Ketamine , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Hippocampus , Ketamine/pharmacology , Ketamine/therapeutic use , Mice , Neurons
3.
Cell Mol Life Sci ; 79(1): 31, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34936033

ABSTRACT

The benefits of current treatments for depression are limited by low response rates, delayed therapeutic effects, and multiple side effects. Antidepressants affect a variety of neurotransmitter systems in different areas of the brain, and the mechanisms underlying their convergent effects on behavior have been unclear. Here we identify hippocampal bone morphogenetic protein (BMP) signaling as a common downstream pathway that mediates the behavioral effects of five different antidepressant classes (fluoxetine, bupropion, duloxetine, vilazodone, trazodone) and of electroconvulsive therapy. All of these therapies decrease BMP signaling and enhance neurogenesis in the hippocampus. Preventing the decrease in BMP signaling blocks the effect of antidepressant treatment on behavioral phenotypes. Further, inhibition of BMP signaling in hippocampal newborn neurons is sufficient to produce an antidepressant effect, while chemogenetic silencing of newborn neurons prevents the antidepressant effect. Thus, inhibition of hippocampal BMP signaling is both necessary and sufficient to mediate the effects of multiple classes of antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Bone Morphogenetic Proteins/metabolism , Hippocampus/metabolism , Signal Transduction , Aging/pathology , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Duloxetine Hydrochloride/pharmacology , Electroconvulsive Therapy , Fluoxetine/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Hippocampus/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Signal Transduction/drug effects , Stress, Psychological/complications , Trazodone/pharmacology , Vilazodone Hydrochloride/pharmacology
4.
Tissue Eng Part A ; 27(3-4): 256-269, 2021 02.
Article in English | MEDLINE | ID: mdl-32580647

ABSTRACT

Stem cell-replacement therapies have been proposed as a potential tool to treat sensorineural hearing loss by aiding the regeneration of spiral ganglion neurons (SGNs) in the inner ear. However, transplantation procedures have yet to be explored thoroughly to ensure proper cell differentiation and optimal transplant procedures. We hypothesized that the aggregation of human embryonic stem cell (hESC)-derived otic neuronal progenitor (ONP) cells into a multicellular form would improve their function and their survival in vivo post-transplantation. We generated hESC-derived ONP spheroids-an aggregate form conducive to differentiation, transplantation, and prolonged cell survival-to optimize conditions for their transplantation. Our findings indicate that these cell spheroids maintain the molecular and functional characteristics similar to those of ONP cells, which are upstream in the SGN lineage. Moreover, our phenotypical, electrophysiological, and mechanical data suggest an optimal spheroid transplantation point after 7 days of in vitro three-dimensional (3D) culture. We have also developed a feasible transplantation protocol for these spheroids using a micropipette aided by a digital microinjection system. In summary, the present work demonstrates that the transplantation of ONP cells in spheroid form into the inner ear through micropipette 7 days after seeding for 3D spheroid culture is an expedient and viable method for stem cell replacement therapies in the inner ear.


Subject(s)
Human Embryonic Stem Cells , Cell Differentiation , Humans , Neurons , Spheroids, Cellular , Spiral Ganglion , Stem Cell Transplantation
5.
Acta Biomater ; 108: 111-127, 2020 05.
Article in English | MEDLINE | ID: mdl-32156626

ABSTRACT

Although the application of human embryonic stem cells (hESCs) in stem cell-replacement therapy remains promising, its potential is hindered by a low cell survival rate in post-transplantation within the inner ear. Here, we aim to enhance the in vitro and in vivo survival rate and neuronal differentiation of otic neuronal progenitors (ONPs) by generating an artificial stem cell niche consisting of three-dimensional (3D) hESC-derived ONP spheroids with a nanofibrillar cellulose hydrogel and a sustained-release brain-derivative neurotrophic factor delivery system. Our results demonstrated that the transplanted hESC-derived ONP spheroids survived and neuronally differentiated into otic neuronal lineages in vitro and in vivo and also extended neurites toward the bony wall of the cochlea 90 days after the transplantation without the use of immunosuppressant medication. Our data in vitro and in vivo presented here provide sufficient evidence that we have established a robust, reproducible protocol for in vivo transplantation of hESC-derived ONPs to the inner ear. Using our protocol to create an artificial stem cell niche in the inner ear, it is now possible to work on integrating transplanted hESC-derived ONPs further and also to work toward achieving functional auditory neurons generated from hESCs. Our findings suggest that the provision of an artificial stem cell niche can be a future approach to stem cell-replacement therapy for inner-ear regeneration. STATEMENT OF SIGNIFICANCE: Inner ear regeneration utilizing human embryonic stem cell-derived otic neuronal progenitors (hESC-derived ONPs) has remarkable potential for treating sensorineural hearing loss. However, the local environment of the inner ear requires a suitable stem cell niche to allow hESC-derived ONP engraftment as well as neuronal differentiation. To overcome this obstacle, we utilized three-dimensional spheroid formation (direct contact), nanofibrillar cellulose hydrogel (extracellular matrix), and a neurotrophic factor delivery system to artificially create a stem cell niche in vitro and in vivo. Our in vitro and in vivo data presented here provide sufficient evidence that we have established a robust, reproducible protocol for in vivo transplantation of hESC-derived ONPs to the inner ear.


Subject(s)
Ear, Inner , Stem Cell Niche , Cell Differentiation , Cellulose , Delayed-Action Preparations , Humans , Hydrogels/pharmacology , Nerve Growth Factors
6.
J Neurotrauma ; 37(3): 494-506, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31516087

ABSTRACT

Gliosis and fibrosis after spinal cord injury (SCI) lead to formation of a scar that is thought to present both molecular and mechanical barriers to neuronal regeneration. The scar consists of a meshwork of reactive glia and deposited, cross-linked, extracellular matrix (ECM) that has long been assumed to present a mechanically "stiff" blockade. However, remarkably little quantitative information is available about the rheological properties of chronically injured spinal tissue. In this study we utilize atomic force microscopy microindentation to provide quantitative evidence of chronic mechanical stiffening after SCI. Using the results of this tissue characterization, we assessed the sensitivity of both mouse and human astrocytes in vitro and determined that they are exquisitely mechanosensitive within the relevant range of substrate stiffness observed in the injured/uninjured spinal cord. We then utilized a novel immune modifying nanoparticle (IMP) treatment as a tool to reveal fibrotic scarring as one of the key drivers of mechanical stiffening after SCI in vivo. We also demonstrate that glial scar-forming astrocytes form a highly aligned, anisotropic network of glial fibers after SCI, and that IMP treatment mitigates this pathological alignment. Taken together, our results identify chronic mechanical stiffening as a critically important aspect of the complex lesion milieu after SCI that must be considered when assessing and developing potential clinical interventions for SCI.


Subject(s)
Biomechanical Phenomena/physiology , Gliosis/physiopathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/physiopathology , Animals , Astrocytes/chemistry , Astrocytes/physiology , Cells, Cultured , Embryonic Stem Cells/chemistry , Embryonic Stem Cells/physiology , Female , Humans , Mice , Mice, Inbred C57BL , Microscopy, Atomic Force/methods , Pregnancy , Thoracic Vertebrae/chemistry
7.
Bone Res ; 7: 33, 2019.
Article in English | MEDLINE | ID: mdl-31700694

ABSTRACT

Heterotopic ossification (HO), true bone formation in soft tissue, is closely associated with abnormal injury/immune responses. We hypothesized that a key underlying mechanism of HO might be injury-induced dysregulation of immune checkpoint proteins (ICs). We found that the earliest stages of HO are characterized by enhanced infiltration of polarized macrophages into sites of minor injuries in an animal model of HO. The non-specific immune suppressants, Rapamycin and Ebselen, prevented HO providing evidence of the central role of the immune responses. We examined the expression pattern of ICs and found that they are dysregulated in HO lesions. More importantly, loss of function of inhibitory ICs (including PD1, PD-L1, and CD152) markedly inhibited HO, whereas loss of function of stimulatory ICs (including CD40L and OX-40L) facilitated HO. These findings suggest that IC inhibitors may provide a therapeutic approach to prevent or limit the extent of HO.

8.
Stem Cell Res Ther ; 10(1): 14, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635039

ABSTRACT

BACKGROUND: Heterotopic ossification (HO), either acquired (aHO) or hereditary, such as fibrodysplasia ossificans progressiva (FOP), is a serious condition without effective treatment. Understanding of the core process of injury-induced HO is still severely limited. METHODS: Double-pulse thymidine analog labeling was used to explore the distinctive domains evolved in injury-induced lesions in an animal model of HO (Nse-BMP4). Histological studies were performed to see whether a similar zonal pattern is also consistently found in biopsies from patients with aHO and FOP. In vivo clonal analysis with Rainbow mice, genetic loss-of-function studies with diphtheria toxin A (DTA)-mediated depletion and lineage tracing with Zsgreen reporter mice were used to obtain further evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells. Immunohistochemistry was used to test whether vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Similar methods also were employed to further understand the signaling pathways that regulate the niche and the resultant HO. RESULTS: We found that distinctive domains evolved in injury-induced lesions, including, from outside-in, a mesenchymal stem cell (MSC) niche, a transient domain and an inner differentiated core in an animal model of HO (Nse-BMP4). A similar zonal structure was found in patients with aHO and FOP. In vivo clonal analysis with Rainbow mice and genetic loss-of-function studies with DTA provided evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells; consistently, vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Further mechanistic study found that BMP and hedgehog (Hh) signaling co-regulate the niche and the resultant HO. CONCLUSIONS: Available data provide evidence of a potential core mechanism in which multiple disease-specific cellular and extracellular molecular elements form a unique local microenvironment, i.e., an injury-induced stem cell niche, which regulates the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs). The implication for HO is that therapeutic approaches must consider several different disease specific factors as parts of a functional unit, instead of treating one factor at a time.


Subject(s)
Myositis Ossificans/genetics , Ossification, Heterotopic/genetics , Osteogenesis/genetics , Stem Cell Niche/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Diphtheria Toxin/genetics , Disease Models, Animal , Excitatory Amino Acid Transporter 1/genetics , Humans , Loss of Function Mutation/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Myositis Ossificans/pathology , Myositis Ossificans/therapy , Ossification, Heterotopic/pathology , Ossification, Heterotopic/therapy , Peptide Fragments/genetics , Receptor, TIE-2/genetics , Signal Transduction/genetics , Zinc Finger Protein GLI1/genetics
9.
Neurobiol Dis ; 116: 60-68, 2018 08.
Article in English | MEDLINE | ID: mdl-29705186

ABSTRACT

Gliosis and fibrosis after spinal cord injury (SCI) lead to formation of a scar that is an impediment to axonal regeneration. Fibrotic scarring is characterized by the accumulation of fibronectin, collagen, and fibroblasts at the lesion site. The mechanisms regulating fibrotic scarring after SCI and its effects on axonal elongation and functional recovery are not well understood. In this study, we examined the effects of eliminating an isoform of fibronectin containing the Extra Domain A domain (FnEDA) on both fibrosis and on functional recovery after contusion SCI using male and female FnEDA-null mice. Eliminating FnEDA did not reduce the acute fibrotic response but markedly diminished chronic fibrotic scarring after SCI. Glial scarring was unchanged after SCI in FnEDA-null mice. We found that FnEDA was important for the long-term stability of the assembled fibronectin matrix during both the subacute and chronic phases of SCI. Motor functional recovery was significantly improved, and there were increased numbers of axons in the lesion site compared to wildtype mice, suggesting that the chronic fibrotic response is detrimental to recovery. Our data provide insight into the mechanisms of fibrosis after SCI and suggest that disruption of fibronectin matrix stability by targeting FnEDA represents a potential therapeutic strategy for promoting recovery after SCI.


Subject(s)
Cicatrix/metabolism , Cicatrix/pathology , Fibronectins/deficiency , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Female , Fibronectins/genetics , Fibrosis/metabolism , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function/physiology
10.
J Neurosci ; 38(15): 3840-3857, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29483282

ABSTRACT

Astrocytes perform a wide array of physiological functions, including structural support, ion exchange, and neurotransmitter uptake. Despite this diversity, molecular markers that label subpopulations of astrocytes are limited, and mechanisms that generate distinct astrocyte subtypes remain unclear. Here we identified serine protease high temperature requirement A 1 (HtrA1), a bone morphogenetic protein 4 signaling regulated protein, as a novel marker of forebrain astrocytes, but not of neural stem cells, in adult mice of both sexes. Genetic deletion of HtrA1 during gliogenesis accelerates astrocyte differentiation. In addition, ablation of HtrA1 in cultured astrocytes leads to altered chondroitin sulfate proteoglycan expression and inhibition of neurite extension, along with elevated levels of transforming growth factor-ß family proteins. Brain injury induces HtrA1 expression in reactive astrocytes, and loss of HtrA1 leads to an impairment in wound closure accompanied by increased proliferation of endothelial and immune cells. Our findings demonstrate that HtrA1 is differentially expressed in adult mouse forebrain astrocytes, and that HtrA1 plays important roles in astrocytic development and injury response.SIGNIFICANCE STATEMENT Astrocytes, an abundant cell type in the brain, perform a wide array of physiological functions. Although characterized as morphologically and functionally diverse, molecular markers that label astrocyte subtypes or signaling pathways that lead to their diversity remain limited. Here, after examining the expression profile of astrocytes generated in response to bone morphogenetic protein signaling, we identify high temperature requirement A 1 (HtrA1) as an astrocyte-specific marker that is differentially expressed in distinct adult mouse brain regions. HtrA1 is a serine protease that has been linked to cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy, a small blood vessel disease in humans. Understanding the role of HtrA1 during development and after injury will provide insights into how distinct astrocyte populations are generated and their unique roles in injury and disease.


Subject(s)
Astrocytes/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Neurogenesis , Wound Healing , Animals , Astrocytes/cytology , Cell Proliferation , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Female , High-Temperature Requirement A Serine Peptidase 1/genetics , Male , Mice , Mice, Inbred C57BL , Prosencephalon/cytology , Transforming Growth Factor beta/metabolism
11.
Bone ; 109: 71-79, 2018 04.
Article in English | MEDLINE | ID: mdl-28645539

ABSTRACT

Heterotopic ossification (HO), acquired or hereditary, endochondral or intramembranous, is the formation of true bone outside the normal skeleton. Since perivascular Gli1+ progenitors contribute to injury induced organ fibrosis, and CD133 is expressed by a variety of populations of adult stem cells, this study utilized Cre-lox based genetic lineage tracing to test the contribution to endochondral HO of adult stem/progenitor cells that expressed either Gli1 or CD133. We found that both lineages contributed broadly to different normal tissues with distinct patterns, but that only Gli1-creERT labeled stem/progenitor cells contributed to all stages of endochondral HO in a BMP dependent, injury induced, transgenic mouse model. Hedgehog (Hh) signaling was abnormal at endochondral HO lesion sites with increased signaling surrounding the lesion but diminished signaling within it. Thus, local dysregulation of Hh signaling participates in the pathophysiology of endochondral HO. However, unlike a previous report of intramembranous HO, systemic inhibition of Hh signaling was insufficient to prevent the initiation of the endochondral HO process or to treat the existing endochondral HO, suggesting that Hh participates in, but is not essential for endochondral HO in this model. This could potentially reflect the underlying difference between intramembranous and endochondral HO. Nevertheless, identification of this novel stem/precursor cell population as a HO-contributing cell population provides a potential drugable target.


Subject(s)
Mesenchymal Stem Cells/metabolism , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Osteogenesis/physiology , Zinc Finger Protein GLI1/metabolism , Animals , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Transgenic , Osteogenesis/genetics , Pyrimidinones/pharmacology , Signal Transduction/genetics , Signal Transduction/physiology , Thiophenes/pharmacology
12.
PLoS One ; 12(12): e0190150, 2017.
Article in English | MEDLINE | ID: mdl-29284013

ABSTRACT

The use of human embryonic stem cells (hESCs) for regeneration of the spiral ganglion will require techniques for promoting otic neuronal progenitor (ONP) differentiation, anchoring of cells to anatomically appropriate and specific niches, and long-term cell survival after transplantation. In this study, we used self-assembling peptide amphiphile (PA) molecules that display an IKVAV epitope (IKVAV-PA) to create a niche for hESC-derived ONPs that supported neuronal differentiation and survival both in vitro and in vivo after transplantation into rodent inner ears. A feature of the IKVAV-PA gel is its ability to form organized nanofibers that promote directed neurite growth. Culture of hESC-derived ONPs in IKVAV-PA gels did not alter cell proliferation or viability. However, the presence of IKVAV-PA gels increased the number of cells expressing the neuronal marker beta-III tubulin and improved neurite extension. The self-assembly properties of the IKVAV-PA gel allowed it to be injected as a liquid into the inner ear to create a biophysical niche for transplanted cells after gelation in vivo. Injection of ONPs combined with IKVAV-PA into the modiolus of X-SCID rats increased survival and localization of the cells around the injection site compared to controls. Human cadaveric temporal bone studies demonstrated the technical feasibility of a transmastoid surgical approach for clinical intracochlear injection of the IKVAV-PA/ONP combination. Combining stem cell transplantation with injection of self-assembling PA gels to create a supportive niche may improve clinical approaches to spiral ganglion regeneration.


Subject(s)
Ear, Inner/metabolism , Peptides/metabolism , Stem Cell Niche , Animals , Cell Differentiation , Cell Transplantation , Cells, Cultured , Ear, Inner/cytology , Humans , Rats
13.
Neurobiol Dis ; 108: 73-82, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823935

ABSTRACT

Intravenously infused synthetic 500nm nanoparticles composed of poly(lactide-co-glycolide) are taken up by blood-borne inflammatory monocytes via a macrophage scavenger receptor (macrophage receptor with collagenous structure), and the monocytes no longer traffic to sites of inflammation. Intravenous administration of the nanoparticles after experimental spinal cord injury in mice safely and selectively limited infiltration of hematogenous monocytes into the injury site. The nanoparticles did not bind to resident microglia, and did not change the number of microglia in the injured spinal cord. Nanoparticle administration reduced M1 macrophage polarization and microglia activation, reduced levels of inflammatory cytokines, and markedly reduced fibrotic scar formation without altering glial scarring. These findings thus implicate early-infiltrating hematogenous monocytes as highly selective contributors to fibrosis that do not play an indispensable role in gliosis after SCI. Further, the nanoparticle treatment reduced accumulation of chondroitin sulfate proteoglycans, increased axon density inside and caudal to the lesion site, and significantly improved functional recovery after both moderate and severe injuries to the spinal cord. These data provide further evidence that hematogenous monocytes contribute to inflammatory damage and fibrotic scar formation after spinal cord injury in mice. Further, since the nanoparticles are simple to administer intravenously, immunologically inert, stable at room temperature, composed of an FDA-approved material, and have no known toxicity, these findings suggest that the nanoparticles potentially offer a practical treatment for human spinal cord injury.


Subject(s)
Immunologic Factors/administration & dosage , Nanoparticles/administration & dosage , Polyglactin 910/administration & dosage , Spinal Cord Injuries/drug therapy , Administration, Intravenous , Animals , Axons/drug effects , Axons/immunology , Axons/pathology , Cell Size , Chondroitin Sulfate Proteoglycans/metabolism , Cicatrix/drug therapy , Cicatrix/immunology , Cicatrix/pathology , Disease Models, Animal , Female , Fibrosis/drug therapy , Fibrosis/immunology , Fibrosis/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Motor Activity/drug effects , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
14.
Stem Cells Transl Med ; 6(3): 923-936, 2017 03.
Article in English | MEDLINE | ID: mdl-28186679

ABSTRACT

The ability to generate spiral ganglion neurons (SGNs) from stem cells is a necessary prerequisite for development of cell-replacement therapies for sensorineural hearing loss. We present a protocol that directs human embryonic stem cells (hESCs) toward a purified population of otic neuronal progenitors (ONPs) and SGN-like cells. Between 82% and 95% of these cells express SGN molecular markers, they preferentially extend neurites to the cochlear nucleus rather than nonauditory nuclei, and they generate action potentials. The protocol follows an in vitro stepwise recapitulation of developmental events inherent to normal differentiation of hESCs into SGNs, resulting in efficient sequential generation of nonneuronal ectoderm, preplacodal ectoderm, early prosensory ONPs, late ONPs, and cells with cellular and molecular characteristics of human SGNs. We thus describe the sequential signaling pathways that generate the early and later lineage species in the human SGN lineage, thereby better describing key developmental processes. The results indicate that our protocol generates cells that closely replicate the phenotypic characteristics of human SGNs, advancing the process of guiding hESCs to states serving inner-ear cell-replacement therapies and possible next-generation hybrid auditory prostheses. © Stem Cells Translational Medicine 2017;6:923-936.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Sensory Receptor Cells/cytology , Spiral Ganglion/cytology , Animals , Brain Stem/cytology , Cell Line , Cell Lineage , Cell Movement , Cell Survival , Coculture Techniques , Human Embryonic Stem Cells/metabolism , Humans , Mice , Neural Stem Cells/cytology , Rats
15.
J Neurosci ; 35(9): 3725-33, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25740503

ABSTRACT

Astrogliosis after spinal cord injury (SCI) is a major impediment to functional recovery. More than half of new astrocytes generated after SCI are derived from ependymal zone stem cells (EZCs). We demonstrate that expression of ß1-integrin increases in EZCs following SCI in mice. Conditional knock-out of ß1-integrin increases GFAP expression and astrocytic differentiation by cultured EZCs without altering oligodendroglial or neuronal differentiation. Ablation of ß1-integrin from EZCs in vivo reduced the number of EZC progeny that continued to express stem cell markers after SCI, increased the proportion of EZC progeny that differentiated into GFAP+ astrocytes, and diminished functional recovery. Loss of ß1-integrin increased SMAD1/5/8 and p38 signaling, suggesting activation of BMP signaling. Coimmunoprecipitation studies demonstrated that ß1-integrin directly interacts with the bone morphogenetic protein receptor subunits BMPR1a and BMPR1b. Ablation of ß1-integrin reduced overall levels of BMP receptors but significantly increased partitioning of BMPR1b into lipid rafts with increased SMAD1/5/8 and p38 signaling. Thus ß1-integrin expression by EZCs reduces movement of BMPR1b into lipid rafts, thereby limiting the known deleterious effects of BMPR1b signaling on glial scar formation after SCI.


Subject(s)
Astrocytes/drug effects , Bone Morphogenetic Protein Receptors/drug effects , Ependyma/cytology , Gliosis/drug therapy , Integrin beta1/pharmacology , Neural Stem Cells/drug effects , Spinal Cord Injuries/drug therapy , Animals , Cell Differentiation , Cells, Cultured , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology
16.
PLoS One ; 9(8): e104335, 2014.
Article in English | MEDLINE | ID: mdl-25098415

ABSTRACT

Astrogliosis with glial scar formation after damage to the nervous system is a major impediment to axonal regeneration and functional recovery. The present study examined the role of ß1-integrin signaling in regulating astrocytic differentiation of neural stem cells. In the adult spinal cord ß1-integrin is expressed predominantly in the ependymal region where ependymal stem cells (ESCs) reside. ß1-integrin signaling suppressed astrocytic differentiation of both cultured ESCs and subventricular zone (SVZ) progenitor cells. Conditional knockout of ß1-integrin enhanced astrogliogenesis both by cultured ESCs and by SVZ progenitor cells. Previous studies have shown that injection into the injured spinal cord of a self-assembling peptide amphiphile that displays an IKVAV epitope (IKVAV-PA) limits glial scar formation and enhances functional recovery. Here we find that injection of IKVAV-PA induced high levels of ß1-integrin in ESCs in vivo, and that conditional knockout of ß1-integrin abolished the astroglial suppressive effects of IKVAV-PA in vitro. Injection into an injured spinal cord of PAs expressing two other epitopes known to interact with ß1-integrin, a Tenascin C epitope and the fibronectin epitope RGD, improved functional recovery comparable to the effects of IKVAV-PA. Finally we found that the effects of ß1-integrin signaling on astrogliosis are mediated by integrin linked kinase (ILK). These observations demonstrate an important role for ß1-integrin/ILK signaling in regulating astrogliosis from ESCs and suggest ILK as a potential target for limiting glial scar formation after nervous system injury.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Integrin beta1/metabolism , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , Astrocytes/cytology , Cell Differentiation/drug effects , Epitopes/pharmacology , Integrin beta1/genetics , Laminin/pharmacology , Mice , Neural Stem Cells/cytology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Long-Evans , Signal Transduction/drug effects
17.
Inflamm Res ; 63(3): 207-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24327087

ABSTRACT

INTRODUCTION: Previous studies found that neuron specific enolase promoter (Nse-BMP4) transgenic mice have increased expression of the nociceptive mediator, substance P and exaggerated local injury responses associated with heterotopic ossification (HO). It is of interest great to know the pain responses in these mice and how the opioid signaling is involved in the downstream events such as mast cell (MC) activation. MATERIALS AND METHODS: This study utilized a transgenic mouse model of HO in which BMP4 is expressed under the control of the Nse-BMP4. The tactile sensitivity and the cold sensitivity of the mice were measured in a classic inflammatory pain model (carrageenan solution injected into the plantar surface of the left hind paw). The MC activation and the expression profiles of different components in the opioid signaling were demonstrated through routine histology and immunohistochemistry and Western blotting, in the superficial and deep muscle injury models. RESULTS: We found that the pain responses in these mice were paradoxically attenuated or unchanged, and we also found increased expression of both Methionine Enkephalin (Met-Enk), and the µ-opioid receptor (MOR). Met-Enk and MOR both co-localized within activated MCs in limb tissues. Further, Nse-BMP4;MOR(-/-) double mutant mice showed attenuated MC activation and had a significant reduction in HO formation in response to injuries. CONCLUSIONS: These observations suggest that opioid signaling may play a key role in MC activation and the downstream inflammatory responses associated with HO. In addition to providing insight into the role of MC activation and associated injury responses in HO, these findings suggest opioid signaling as a potential therapeutic target in HO and possibly others disorders involving MC activation.


Subject(s)
Enkephalin, Methionine/physiology , Mast Cells/physiology , Ossification, Heterotopic/physiopathology , Animals , Bone Morphogenetic Protein 4/genetics , Cold Temperature , Immunohistochemistry , Inflammation/complications , Inflammation/pathology , Mast Cells/pathology , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/injuries , Mutation/genetics , Mutation/physiology , Nociception/physiology , Ossification, Heterotopic/pathology , Pain Measurement , Phosphopyruvate Hydratase/genetics , Physical Stimulation , Receptors, Opioid, mu/physiology , Signal Transduction/physiology
18.
Bone ; 53(1): 194-203, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262027

ABSTRACT

Heterotopic ossification (HO), acquired or hereditary, is the formation of true bone outside the normal skeleton. Although the lineages of cells contributing to bone formation during normal development are well defined, the precise lineages of cells that contribute to HO are not clear. This study utilized Cre-lox based genetic lineage tracing to examine the contribution to HO of cells that expressed either FoxD1 or Glast. Both lineages contributed broadly to different normal tissues, and FoxD1-cre labeled cells contributed to normal bone formation. Despite the similarity in labeling patterns of normal tissues, and the significant contribution of FoxD1-cre labeled cells to normal bone, only Glast-creERT labeled progenitors contributed significantly to HO at all stages, suggesting that the cell populations that normally contribute to physiological bone formation, such as the Foxd1-cre labeled cells, may not participate in pathological HO. Further, identification of Glast-expressing cells as precursors that give rise to HO should help with the molecular targeting of this population both for the prevention and for the treatment of HO.


Subject(s)
Ossification, Heterotopic , Stem Cells/pathology , Animals , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Recombination, Genetic
19.
J Neurosci ; 32(50): 17935-47, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23238710

ABSTRACT

Astrogliosis following spinal cord injury (SCI) involves an early hypertrophic response that serves to repair damaged blood-brain barrier and a subsequent hyperplastic response that results in a dense scar that impedes axon regeneration. The mechanisms regulating these two phases of astrogliosis are beginning to be elucidated. In this study, we found that microRNA-21 (miR-21) increases in a time-dependent manner following SCI in mouse. Astrocytes adjacent to the lesion area express high levels of miR-21 whereas astrocytes in uninjured spinal cord express low levels of miR-21. To study the role of miR-21 in astrocytes after SCI, transgenic mice were generated that conditionally overexpress either the primary miR-21 transcript in astrocytes or a miRNA sponge designed to inhibit miR-21 function. Overexpression of miR-21 in astrocytes attenuated the hypertrophic response to SCI. Conversely, expression of the miR-21 sponge augmented the hypertrophic phenotype, even in chronic stages of SCI recovery when astrocytes have normally become smaller in size with fine processes. Inhibition of miR-21 function in astrocytes also resulted in increased axon density within the lesion site. These findings demonstrate a novel role for miR-21 in regulating astrocytic hypertrophy and glial scar progression after SCI, and suggest miR-21 as a potential therapeutic target for manipulating gliosis and enhancing functional outcome.


Subject(s)
Astrocytes/metabolism , MicroRNAs/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Animals , Disease Models, Animal , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/pathology
20.
J Neurol ; 259(12): 2644-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22736080

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder of progressive heterotopic ossification (HO) caused by a recurrent activating mutation of ACVR1/ALK2, a bone morphogenetic protein (BMP) type I receptor. FOP is characterized by progressive HO, which is associated with inflammation in the setting of dysregulated BMP signaling, however, a variety of atypical neurologic symptoms are also reported by FOP patients. The main objective of this study is to investigate the potential underlying mechanism that is responsible for the observed atypical neurologic symptoms. We evaluated two mouse models of dysregulated BMP signaling for potential CNS pathology through non-invasive magnetic resonance imaging (MRI) studies and histological and immunohistochemical approaches. In one model, BMP4 is over-expressed under the control of the neuron-specific enolase promoter; the second model is a knock-in of a recurrent FOP mutation of ACVR1/ALK2. We also retrospectively examined MRI scans of four FOP patients. We consistently observed demyelinated lesions and focal inflammatory changes of the CNS in both mouse models but not in wild-type controls, and also found CNS white matter lesions in each of the four FOP patients examined. These findings suggest that dysregulated BMP signaling disturbs normal homeostasis of target tissues, including CNS where focal demyelination may manifest as the neurologic symptoms frequently observed in FOP.


Subject(s)
Demyelinating Diseases/complications , Demyelinating Diseases/diagnosis , Myositis Ossificans/complications , Myositis Ossificans/diagnosis , Adolescent , Adult , Animals , Child, Preschool , Demyelinating Diseases/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Myositis Ossificans/genetics , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...