1
|
Chen L, Yang J, Klassen H. Immune Responses to Sequential Binocular Transplantation of Allogeneic Retinal Progenitor Cells to the Vitreous Cavity in Mice. Int J Mol Sci 2023; 24:ijms24076205. [PMID: 37047179 PMCID: PMC10093920 DOI: 10.3390/ijms24076205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Intravitreal transplantation of allogeneic human retinal progenitor cells (hRPCs) holds promise as a treatment for blinding retinal degenerations. Prior work has shown that neural progenitors are well-tolerated as allografts following single injections; however, sequential delivery of allogeneic cells raises the potential risk of host sensitization with subsequent immune rejection of grafts. The current study was designed to assess whether an immune response would be induced by repeated intravitreal transplants of allogeneic RPCs utilizing the mouse animal model. We injected murine retinal progenitor cells (gmRPCs), originally derived from donors with a C57BL/6 genetic background, into BALB/c recipient mice in order to provide safety data as to what might be expected following repeated treatment of patients with allogeneic human cell product. Immune responses to gmRPCs were mild, consisting of T cells, B cells, neutrophils, and natural killer cells, with macrophages clearly the predominating. Animals treated with repeat doses of gmRPCs did not show evidence of sensitization, nor was there immune-mediated destruction of the grafts. Despite the absence of immunosuppressive treatments, allogeneic gmRPC grafts survived following repeat dosing, thus providing support for the preliminary observation that repeated injection of allogeneic RPCs to the vitreous cavity is tolerated in patients with retinitis pigmentosa.
Collapse
|
2
|
Var SR, Strell P, Johnson ST, Roman A, Vasilakos Z, Low WC. Transplanting Microglia for Treating CNS Injuries and Neurological Diseases and Disorders, and Prospects for Generating Exogenic Microglia. Cell Transplant 2023; 32:9636897231171001. [PMID: 37254858 PMCID: PMC10236244 DOI: 10.1177/09636897231171001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/18/2023] [Accepted: 04/05/2023] [Indexed: 06/01/2023] Open
Abstract
Microglia are associated with a wide range of both neuroprotective and neuroinflammatory functions in the central nervous system (CNS) during development and throughout lifespan. Chronically activated and dysfunctional microglia are found in many diseases and disorders, such as Alzheimer's disease, Parkinson's disease, and CNS-related injuries, and can accelerate or worsen the condition. Transplantation studies designed to replace and supplement dysfunctional microglia with healthy microglia offer a promising strategy for addressing microglia-mediated neuroinflammation and pathologies. This review will cover microglial involvement in neurological diseases and disorders and CNS-related injuries, current microglial transplantation strategies, and different approaches and considerations for generating exogenic microglia.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, Medical
School, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, Medical School,
University of Minnesota, Minneapolis, MN, USA
| | - Phoebe Strell
- Stem Cell Institute, Medical School,
University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary and Biomedical
Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Sether T. Johnson
- Department of Neurosurgery, Medical
School, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, Medical School,
University of Minnesota, Minneapolis, MN, USA
| | - Alex Roman
- Department of Neuroscience, University
of Minnesota, Minneapolis, MN, USA
| | - Zoey Vasilakos
- Stem Cell Institute, Medical School,
University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University
of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Department of Neurosurgery, Medical
School, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, Medical School,
University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary and Biomedical
Sciences, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
3
|
Satterlee AB, Dunn DE, Valdivia A, Malawsky D, Buckley A, Gershon T, Floyd S, Hingtgen S. Spatiotemporal analysis of induced neural stem cell therapy to overcome advanced glioblastoma recurrence. Mol Ther Oncolytics 2022; 26:49-62. [PMID: 35784402 PMCID: PMC9217992 DOI: 10.1016/j.omto.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/01/2022] [Indexed: 12/03/2022] Open
Abstract
Genetically engineered neural stem cells (NSCs) are a promising therapy for the highly aggressive brain cancer glioblastoma (GBM); however, treatment durability remains a major challenge. We sought to define the events that contribute to dynamic adaptation of GBM during treatment with human skin-derived induced NSCs releasing the pro-apoptotic agent TRAIL (iNSC-TRAIL) and develop strategies that convert initial tumor kill into sustained GBM suppression. In vivo and ex vivo analysis before, during, and after treatment revealed significant shifts in tumor transcriptome and spatial distribution as the tumors adapted to treatment. To address this, we designed iNSC delivery strategies that increased spatiotemporal TRAIL coverage and significantly decreased GBM volume throughout the brain, reducing tumor burden 100-fold as quantified in live ex vivo brain slices. The varying impact of different strategies on treatment durability and median survival of both solid and invasive tumors provides important guidance for optimizing iNSC therapy.
Collapse
Affiliation(s)
- Andrew B. Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Denise E. Dunn
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27704, USA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Daniel Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Andrew Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Timothy Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Scott Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27704, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Smith MJ, Paton MCB, Fahey MC, Jenkin G, Miller SL, Finch-Edmondson M, McDonald CA. Neural stem cell treatment for perinatal brain injury: A systematic review and meta-analysis of preclinical studies. Stem Cells Transl Med 2021; 10:1621-1636. [PMID: 34542242 PMCID: PMC8641092 DOI: 10.1002/sctm.21-0243] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 12/15/2022] Open
Abstract
Perinatal brain injury can lead to significant neurological and cognitive deficits and currently no therapies can regenerate the damaged brain. Neural stem cells (NSCs) have the potential to engraft and regenerate damaged brain tissue. The aim of this systematic review was to evaluate the preclinical literature to determine whether NSC administration is more effective than controls in decreasing perinatal brain injury. Controlled interventional studies of NSC therapy using animal models of perinatal brain injury were identified using MEDLINE and Embase. Primary outcomes were brain infarct size, motor, and cognitive function. Data for meta‐analysis were synthesized and expressed as standardized mean difference (SMD) with 95% confidence intervals (CI), using a random effects model. We also reported secondary outcomes including NSC survival, migration, differentiation, and effect on neuroinflammation. Eighteen studies met inclusion criteria. NSC administration decreased infarct size (SMD 1.09; CI: 0.44, 1.74, P = .001; I2 = 74%) improved motor function measured via the impaired forelimb preference test (SMD 2.27; CI: 0.85, 3.69, P = .002; I2 = 86%) and the rotarod test (SMD 1.88; CI: 0.09, 3.67, P = .04; I2 = 95%). Additionally, NSCs improved cognitive function measured via the Morris water maze test (SMD of 2.41; CI: 1.16, 3.66, P = .0002; I2 = 81%). Preclinical evidence suggests that NSC therapy is promising for the treatment of perinatal brain injury. We have identified key knowledge gaps, including the lack of large animal studies and uncertainty regarding the necessity of immunosuppression for NSC transplantation in neonates. These knowledge gaps should be addressed before NSC treatment can effectively progress to clinical trial.
Collapse
Affiliation(s)
- Madeleine J Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Madison Claire Badawy Paton
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Megan Finch-Edmondson
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Yang H, Wang F, Liu X, Wang H, Qu T. Mesenchymal stem cells from human umbilical cord regulate the expression of major histocompatibility complex in human neural stem cells and their lineages. Neurosci Lett 2020; 738:135359. [PMID: 32949939 DOI: 10.1016/j.neulet.2020.135359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/13/2020] [Accepted: 09/02/2020] [Indexed: 12/31/2022]
Abstract
hNSCs (human neural stem cells) derived from embryonic tissue and aborted fetal brains are considered to be the most promising candidates for neurodegenerative and other CNS(central nervous system) diseases. However, the most common problem, which limited successful use of these allogeneic hNSC therapy, is immune rejection. Mesenchymal stem cells (MSCs) from human umbilical cord (hUC-MSCs) are receiving increasing attention for their immune-modulatory properties. In the current studies, we firstly investigated the immunogenecity of hNSCs as well as their lineages in cultures with the presence or absence of interferon gamma (IFNγ), a pro-inflammatory factors. Our data revealed that the majority of hNSCs and astrocytes expressed MHCI (major histocompatibility complex class I) while neurons hardly expressed MHCI (<5%) in the absence of IFNγ. In addition, neither hNSCs nor neurons expressed MHCII while a subpopulation (about 18 %) of astrocytes expressed MHCII without IFNγ stimulation. However, the addition of IFNγ in cultures significantly increased the expressions of MHCII on hNSCs and astrocytes. However, IFNγ did not affect the expression of MHCI on hNSCs and astrocytes. We then investigated whether hUC-MSCs had the capacity of regulating the immunogenecity of hNSCs as well as their lineages in a co-culture system. We found that hUC-MSCs did not affect the expression of MHCI on hNSCs and their lineages, however, these cells were able to significantly inhibit the IFNγ-induced up-regulation of MHCII on hNSCs and astrocytes (p < 0.001). Thus, our results suggest that hUC-MSCs may serve as potentially useful modulators to reduce the immunogenicity of allogeneic hNSCs in clinical application.
Collapse
Affiliation(s)
- Hongna Yang
- Department of Critical-care Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, 250012, China.
| | - Feng Wang
- Department of Breast Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaodun Liu
- R & D of Cell and Tissue Bank, Qilu Stem Cell Engineering Company of Shandong Province, Jinan, Shandong, 250000, China
| | - Hao Wang
- Department of Critical-care Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, 250012, China
| | - Tingyu Qu
- R & D of Cell and Tissue Bank, Qilu Stem Cell Engineering Company of Shandong Province, Jinan, Shandong, 250000, China.
| |
Collapse
|
6
|
Ottoboni L, von Wunster B, Martino G. Therapeutic Plasticity of Neural Stem Cells. Front Neurol 2020; 11:148. [PMID: 32265815 PMCID: PMC7100551 DOI: 10.3389/fneur.2020.00148] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have garnered significant scientific and commercial interest in the last 15 years. Given their plasticity, defined as the ability to develop into different phenotypes inside and outside of the nervous system, with a capacity of almost unlimited self-renewal, of releasing trophic and immunomodulatory factors, and of exploiting temporal and spatial dynamics, NSCs have been proposed for (i) neurotoxicity testing; (ii) cellular therapies to treat CNS diseases; (iii) neural tissue engineering and repair; (iv) drug target validation and testing; (v) personalized medicine. Moreover, given the growing interest in developing cell-based therapies to target neurodegenerative diseases, recent progress in developing NSCs from human-induced pluripotent stem cells has produced an analog of endogenous NSCs. Herein, we will review the current understanding on emerging conceptual and technological topics in the neural stem cell field, such as deep characterization of the human compartment, single-cell spatial-temporal dynamics, reprogramming from somatic cells, and NSC manipulation and monitoring. Together, these aspects contribute to further disentangling NSC plasticity to better exploit the potential of those cells, which, in the future, might offer new strategies for brain therapies.
Collapse
Affiliation(s)
- Linda Ottoboni
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Gianvito Martino
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, School of Medicine, Milan, Italy
| |
Collapse
|
7
|
Mangale V, McIntyre LL, Walsh CM, Loring JF, Lane TE. Promoting remyelination through cell transplantation therapies in a model of viral-induced neurodegenerative disease. Dev Dyn 2018; 248:43-52. [PMID: 30067309 DOI: 10.1002/dvdy.24658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) disease characterized by chronic neuroinflammation, demyelination, and axonal damage. Infiltration of activated lymphocytes and myeloid cells are thought to be primarily responsible for white matter damage and axonopathy. Several United States Food and Drug Administration-approved therapies exist that impede activated lymphocytes from entering the CNS thereby limiting new lesion formation in patients with relapse-remitting forms of MS. However, a significant challenge within the field of MS research is to develop effective and sustained therapies that allow for axonal protection and remyelination. In recent years, there has been increasing evidence that some kinds of stem cells and their derivatives seem to be able to mute neuroinflammation as well as promote remyelination and axonal integrity. Intracranial infection of mice with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in immune-mediated demyelination and axonopathy, making this an excellent model to interrogate the therapeutic potential of stem cell derivatives in evoking remyelination. This review provides a succinct overview of our recent findings using intraspinal injection of mouse CNS neural progenitor cells and human neural precursors into JHMV-infected mice. JHMV-infected mice receiving these cells display extensive remyelination associated with axonal sparing. In addition, we discuss possible mechanisms associated with sustained clinical recovery. Developmental Dynamics 248:43-52, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vrushali Mangale
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Laura L McIntyre
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Center, University of California, Irvine, California
| | - Craig M Walsh
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Center, University of California, Irvine, California
| | - Jeanne F Loring
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Thomas E Lane
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Department of Bioengineering, University of Utah, Salt Lake City, Utah.,Immunology, Inflammation, and Infectious Disease Initiative, University of Utah, Salt Lake City, Utah
| |
Collapse
|
8
|
Mesenchymal Stem Cells Form 3D Clusters Following Intraventricular Transplantation. J Mol Neurosci 2018; 65:60-73. [PMID: 29705933 DOI: 10.1007/s12031-018-1070-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are regarded as an immune privileged cell type with numerous regeneration-promoting effects. The in vivo behavior of MSC and underlying mechanisms leading to their regenerative effects are largely unknown. The aims of this study were to comparatively investigate the in vivo behavior of canine (cMSC), human (hMSC), and murine MSC (mMSC) following intra-cerebroventricular transplantation. At 7 days post transplantation (dpt), clusters of cMSC, hMSC, and mMSC were detected within the ventricular system. At 49 dpt, cMSC-transplanted mice showed clusters mostly consisting of extracellular matrix lacking transplanted MSC. Similarly, hMSC-transplanted mice lacked MSC clusters at 49 dpt. Xenogeneic MSC transplantation was associated with a local T lymphocyte-dominated immune reaction at both time points. Interestingly, no associated inflammation was observed following syngeneic mMSC transplantation. In conclusion, transplanted MSC formed intraventricular cell clusters and exhibited a short life span in vivo. Xenogeneically in contrast to syngeneically transplanted MSC triggered a T cell-mediated graft rejection indicating that MSCs are not as immune privileged as previously assumed. However, MSC may mediate their effects by a "hit and run" mechanism and future studies will show whether syngeneically or xenogeneically transplanted MSCs exert better therapeutic effects in animals with CNS disease.
Collapse
|
9
|
Varrault A, Eckardt S, Girard B, Le Digarcher A, Sassetti I, Meusnier C, Ripoll C, Badalyan A, Bertaso F, McLaughlin KJ, Journot L, Bouschet T. Mouse Parthenogenetic Embryonic Stem Cells with Biparental-Like Expression of Imprinted Genes Generate Cortical-Like Neurons That Integrate into the Injured Adult Cerebral Cortex. Stem Cells 2017; 36:192-205. [PMID: 29044892 DOI: 10.1002/stem.2721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 09/19/2017] [Accepted: 10/07/2017] [Indexed: 01/10/2023]
Abstract
One strategy for stem cell-based therapy of the cerebral cortex involves the generation and transplantation of functional, histocompatible cortical-like neurons from embryonic stem cells (ESCs). Diploid parthenogenetic Pg-ESCs have recently emerged as a promising source of histocompatible ESC derivatives for organ regeneration but their utility for cerebral cortex therapy is unknown. A major concern with Pg-ESCs is genomic imprinting. In contrast with biparental Bp-ESCs derived from fertilized oocytes, Pg-ESCs harbor two maternal genomes but no sperm-derived genome. Pg-ESCs are therefore expected to have aberrant expression levels of maternally expressed (MEGs) and paternally expressed (PEGs) imprinted genes. Given the roles of imprinted genes in brain development, tissue homeostasis and cancer, their deregulation in Pg-ESCs might be incompatible with therapy. Here, we report that, unexpectedly, only one gene out of 7 MEGs and 12 PEGs was differentially expressed between Pg-ESCs and Bp-ESCs while 13 were differentially expressed between androgenetic Ag-ESCs and Bp-ESCs, indicating that Pg-ESCs but not Ag-ESCs, have a Bp-like imprinting compatible with therapy. In vitro, Pg-ESCs generated cortical-like progenitors and electrophysiologically active glutamatergic neurons that maintained the Bp-like expression levels for most imprinted genes. In vivo, Pg-ESCs participated to the cortical lineage in fetal chimeras. Finally, transplanted Pg-ESC derivatives integrated into the injured adult cortex and sent axonal projections in the host brain. In conclusion, mouse Pg-ESCs generate functional cortical-like neurons with Bp-like imprinting and their derivatives properly integrate into both the embryonic cortex and the injured adult cortex. Collectively, our data support the utility of Pg-ESCs for cortical therapy. Stem Cells 2018;36:192-205.
Collapse
Affiliation(s)
- Annie Varrault
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Sigrid Eckardt
- Research Institute at Nationwide Children's Hospital, Center for Molecular and Human Genetics, Columbus, Ohio, USA
| | - Benoît Girard
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Anne Le Digarcher
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Isabelle Sassetti
- Institute for Neuroscience of Montpellier, Hôpital Saint Eloi, Montpellier cedex 5, France
| | - Céline Meusnier
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Chantal Ripoll
- Institute for Neuroscience of Montpellier, Hôpital Saint Eloi, Montpellier cedex 5, France
| | - Armen Badalyan
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Federica Bertaso
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - K John McLaughlin
- Research Institute at Nationwide Children's Hospital, Center for Molecular and Human Genetics, Columbus, Ohio, USA
| | - Laurent Journot
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Tristan Bouschet
- Institut de Génomique Fonctionnelle, IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| |
Collapse
|
10
|
Mangale V, Marro BS, Plaisted WC, Walsh CM, Lane TE. Neural precursor cells derived from induced pluripotent stem cells exhibit reduced susceptibility to infection with a neurotropic coronavirus. Virology 2017; 511:49-55. [PMID: 28822268 PMCID: PMC5623645 DOI: 10.1016/j.virol.2017.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/28/2017] [Accepted: 08/02/2017] [Indexed: 12/17/2022]
Abstract
The present study examines the susceptibility of mouse induced pluripotent stem cell-derived neural precursor cells (iPSC-NPCs) to infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Similar to NPCs derived from striatum of day 1 postnatal GFP-transgenic mice (GFP-NPCs), iPSC-derived NPCs (iPSC-NPCs) are able to differentiate into terminal neural cell types and express MHC class I and II in response to IFN-γ treatment. However, in contrast to postnatally-derived NPCs, iPSC-NPCs express low levels of carcinoembryonic antigen-cell adhesion molecule 1a (CEACAM1a), the surface receptor for JHMV, and are less susceptible to infection and virus-induced cytopathic effects. The relevance of this in terms of therapeutic application of NPCs resistant to viral infection is discussed. The neurotropic virus JHMV infects and kills mouse post-natal neural progenitor cells (NPCs). This study examines if JHMV infects mouse inducible pluripotent stem cell-derived NPCs. iPSC-NPCs are less susceptible to infection with JHMV and subsequent lytic effects.
Collapse
Affiliation(s)
- Vrushali Mangale
- Department of Pathology, Division of Microbiology & Immunology University of Utah, Salt Lake City, UT 84112, United States
| | - Brett S Marro
- Department of Molecular Biology & Biochemistry and Institute for Immunology, University of California, Irvine 92697, United States
| | - Warren C Plaisted
- Department of Molecular Biology & Biochemistry and Institute for Immunology, University of California, Irvine 92697, United States
| | - Craig M Walsh
- Department of Molecular Biology & Biochemistry and Institute for Immunology, University of California, Irvine 92697, United States
| | - Thomas E Lane
- Department of Pathology, Division of Microbiology & Immunology University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
11
|
Tanabe T, Watanabe H, Shah JA, Sahara H, Shimizu A, Nomura S, Asfour A, Danton M, Boyd L, Meyers AD, Ekanayake-Alper DK, Sachs DH, Yamada K. Role of Intrinsic (Graft) Versus Extrinsic (Host) Factors in the Growth of Transplanted Organs Following Allogeneic and Xenogeneic Transplantation. Am J Transplant 2017; 17:1778-1790. [PMID: 28117931 PMCID: PMC5489354 DOI: 10.1111/ajt.14210] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/08/2016] [Accepted: 01/09/2017] [Indexed: 01/25/2023]
Abstract
In our studies of life-supporting α-1,3-galactocyltransferase knockout (GalT-KO) pig-to-baboon kidneys, we found that some recipients developed increased serum creatinine with growth of the grafts, without histological or immunological evidence of rejection. We hypothesized that the rapid growth of orthotopic pig grafts in smaller baboon recipients may have led to deterioration of organ function. To test this hypothesis for both kidneys and lungs, we assessed whether the growth of outbred (Yorkshire) organ transplants in miniature swine was regulated by intrinsic (graft) or extrinsic (host environment) factors. Yorkshire kidneys exhibited persistent growth in miniature swine, reaching 3.7 times their initial volume over 3 mo versus 1.2 times for miniature swine kidneys over the same time period. Similar rapid early growth of lung allografts was observed and, in this case, led to organ dysfunction. For xenograft kidneys, a review of our results suggests that there is a threshold for kidney graft volume of 25 cm3 /kg of recipient body weight at which cortical ischemia is induced in transplanted GalT-KO kidneys in baboons. These results suggest that intrinsic factors are responsible, at least in part, for growth of donor organs and that this property should be taken into consideration for growth-curve-mismatched transplants, especially for life-supporting organs transplanted into a limited recipient space.
Collapse
Affiliation(s)
- Tatsu Tanabe
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY,Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Hironosuke Watanabe
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Jigesh A Shah
- Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Hisashi Sahara
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY,Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shunichiro Nomura
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Arsenoi Asfour
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Makenzie Danton
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Lennan Boyd
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY
| | - Adrienne Dardenne Meyers
- Skirball Center for Cardiovascular Research, Cardiovascular Research Foundation, Orangeburg, New York
| | | | - David H Sachs
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY,Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Kazuhiko Yamada
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY,Transplantation Biology Research Center Laboratory, Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA,Corresponding author: Kazuhiko Yamada, M.D, PhD., Columbia Center for Translational Immunology, 630 W 168th St, BB1705, New York, NY, USA, Tel: +1-212-304-5695,
| |
Collapse
|
12
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
13
|
Plaisted WC, Zavala A, Hingco E, Tran H, Coleman R, Lane TE, Loring JF, Walsh CM. Remyelination Is Correlated with Regulatory T Cell Induction Following Human Embryoid Body-Derived Neural Precursor Cell Transplantation in a Viral Model of Multiple Sclerosis. PLoS One 2016; 11:e0157620. [PMID: 27310015 PMCID: PMC4911106 DOI: 10.1371/journal.pone.0157620] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 06/02/2016] [Indexed: 02/06/2023] Open
Abstract
We have recently described sustained clinical recovery associated with dampened neuroinflammation and remyelination following transplantation of neural precursor cells (NPCs) derived from human embryonic stem cells (hESCs) in a viral model of the human demyelinating disease multiple sclerosis. The hNPCs used in that study were derived by a novel direct differentiation method (direct differentiation, DD-NPCs) that resulted in a unique gene expression pattern when compared to hNPCs derived by conventional methods. Since the therapeutic potential of human NPCs may differ greatly depending on the method of derivation and culture, we wanted to determine whether NPCs differentiated using conventional methods would be similarly effective in improving clinical outcome under neuroinflammatory demyelinating conditions. For the current study, we utilized hNPCs differentiated from a human induced pluripotent cell line via an embryoid body intermediate stage (EB-NPCs). Intraspinal transplantation of EB-NPCs into mice infected with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in decreased accumulation of CD4+ T cells in the central nervous system that was concomitant with reduced demyelination at the site of injection. Dampened neuroinflammation and remyelination was correlated with a transient increase in CD4+FOXP3+ regulatory T cells (Tregs) concentrated within the peripheral lymphatics. However, compared to our earlier study, pathological improvements were modest and did not result in significant clinical recovery. We conclude that the genetic signature of NPCs is critical to their effectiveness in this model of viral-induced neurologic disease. These comparisons will be useful for understanding what factors are critical for the sustained clinical improvement.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- Cell Differentiation
- Cell- and Tissue-Based Therapy/methods
- Coronavirus Infections/immunology
- Coronavirus Infections/pathology
- Coronavirus Infections/therapy
- Coronavirus Infections/virology
- Disease Models, Animal
- Embryoid Bodies/cytology
- Embryoid Bodies/immunology
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression
- Hepatitis, Viral, Animal/immunology
- Hepatitis, Viral, Animal/pathology
- Hepatitis, Viral, Animal/therapy
- Hepatitis, Viral, Animal/virology
- Human Embryonic Stem Cells/cytology
- Human Embryonic Stem Cells/immunology
- Humans
- Lymphocyte Activation
- Male
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/therapy
- Murine hepatitis virus/growth & development
- Murine hepatitis virus/pathogenicity
- Myelin Sheath/immunology
- Neural Stem Cells/cytology
- Neural Stem Cells/immunology
- Neural Stem Cells/transplantation
- Organ Specificity
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Warren C. Plaisted
- Department of Molecular Biology & Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, Institute for Immunology, University of California Irvine, Irvine, California, United States of America
| | - Angel Zavala
- Department of Molecular Biology & Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, Institute for Immunology, University of California Irvine, Irvine, California, United States of America
| | - Edna Hingco
- Department of Molecular Biology & Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, Institute for Immunology, University of California Irvine, Irvine, California, United States of America
| | - Ha Tran
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ronald Coleman
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas E. Lane
- Department of Pathology, University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail: (CMW); (JFL); (TEL)
| | - Jeanne F. Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail: (CMW); (JFL); (TEL)
| | - Craig M. Walsh
- Department of Molecular Biology & Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, Institute for Immunology, University of California Irvine, Irvine, California, United States of America
- * E-mail: (CMW); (JFL); (TEL)
| |
Collapse
|
14
|
Führmann T, Tam R, Ballarin B, Coles B, Elliott Donaghue I, van der Kooy D, Nagy A, Tator C, Morshead C, Shoichet M. Injectable hydrogel promotes early survival of induced pluripotent stem cell-derived oligodendrocytes and attenuates longterm teratoma formation in a spinal cord injury model. Biomaterials 2016; 83:23-36. [DOI: 10.1016/j.biomaterials.2015.12.032] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/14/2015] [Accepted: 12/29/2015] [Indexed: 02/06/2023]
|
15
|
Ottoboni L, De Feo D, Merlini A, Martino G. Commonalities in immune modulation between mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs). Immunol Lett 2015; 168:228-39. [DOI: 10.1016/j.imlet.2015.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
|
16
|
Scutellarin Alleviates Behavioral Deficits in a Mouse Model of Multiple Sclerosis, Possibly Through Protecting Neural Stem Cells. J Mol Neurosci 2015; 58:210-20. [PMID: 26514969 DOI: 10.1007/s12031-015-0660-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/24/2015] [Indexed: 12/14/2022]
Abstract
Scutellarin, a flavonoid extracted from an herbal medication (Erigeron breviscapus Hand-Mazz), has been shown to protect neurons against damage and to promote neurogenesis, and thus has therapeutic potential in the treatment of a variety of neurodegenerative diseases. Since neural stem cells (NSCs) could differentiate into myelin-producing oligodendrocytes, we speculate that scutellarin could also be used to treat multiple sclerosis (MS). In the current study, we examined potential effects of scutellarin using a mouse model of MS. Briefly, adult C57BL/6 mice exposed to cuprizone (8 mg/day through diet, for 6 consecutive weeks) randomly received scutellarin (50 mg/kg/day) or vehicle for 10 consecutive days. In the scutellarin-treated group, rotarod testing at the end of the treatment showed significant improvement of motor function (increased time to fall); myelin basic protein (MBP) staining of the corpus callosum revealed decreased demyelination; TUNEL staining followed by Nestin or Sox2 staining revealed increased number of NSCs and decreased rate of NSC apoptosis in the subventricular zone (SVZ) of the lateral ventricles (LV). In a series of experiments using cultured NSCs subjected to cuprizone injury, we confirmed the protective effects of scutellarin. At 30 μM, scutellarin increased the commitment of NSCs to the oligodendrocyte and neuronal lineages, as evidenced by NG2 chondroitin sulfate proteoglycan (NG2) and doublecortin (DCX) staining. Differentiation into astrocytes (as revealed by glial fibrillary acidic protein (GFAP) staining) was decreased. Maturation of the NSCs committed to the oligodendrocyte lineage, as evidenced by oligodendrocyte marker O4 antibody (O4) staining and MBP staining, was also promoted by scutellarin. Further analysis revealed that scutellarin might suppress the phosphorylation of p38 in cuprizone-induced NSCs. In summary, scutellarin could alleviate motor deficits in a mouse model for MS, possibly by inhibiting NSC apoptosis and promoting differentiation of NSCs to myelin-producing oligodendrocytes.
Collapse
|
17
|
Boltze J, Arnold A, Walczak P, Jolkkonen J, Cui L, Wagner DC. The Dark Side of the Force - Constraints and Complications of Cell Therapies for Stroke. Front Neurol 2015; 6:155. [PMID: 26257702 PMCID: PMC4507146 DOI: 10.3389/fneur.2015.00155] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/23/2015] [Indexed: 12/16/2022] Open
Abstract
Cell therapies are increasingly recognized as a promising option to augment the limited therapeutic arsenal available to fight ischemic stroke. During the last two decades, cumulating preclinical evidence has indicated a substantial efficacy for most cell treatment paradigms and first clinical trials are currently underway to assess safety and feasibility in patients. However, the strong and still unmet demand for novel stroke treatment options and exciting findings reported from experimental studies may have drawn our attention away from potential side effects related to cell therapies and the ways by which they are commonly applied. This review summarizes common and less frequent adverse events that have been discovered in preclinical and clinical investigations assessing cell therapies for stroke. Such adverse events range from immunological and neoplastic complications over seizures to cell clotting and cell-induced embolism. It also describes potential complications of clinically applicable administration procedures, detrimental interactions between therapeutic cells, and the pathophysiological environment that they are placed into, as well as problems related to cell manufacturing. Virtually each therapeutic intervention comes at a certain risk for complications. Side effects do therefore not generally compromise the value of cell treatments for stroke, but underestimating such complications might severely limit therapeutic safety and efficacy of cell treatment protocols currently under development. On the other hand, a better understanding will provide opportunities to further improve existing therapeutic strategies and might help to define those circumstances, under which an optimal effect can be realized. Hence, the review eventually discusses strategies and recommendations allowing us to prevent or at least balance potential complications in order to ensure the maximum therapeutic benefit at minimum risk for stroke patients.
Collapse
Affiliation(s)
- Johannes Boltze
- Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology , Leipzig , Germany ; Translational Center for Regenerative Medicine, University of Leipzig , Leipzig , Germany
| | - Antje Arnold
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Institute for Cell Engineering, Johns Hopkins University , Baltimore, MD , USA
| | - Piotr Walczak
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Institute for Cell Engineering, Johns Hopkins University , Baltimore, MD , USA
| | - Jukka Jolkkonen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Lili Cui
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Daniel-Christoph Wagner
- Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology , Leipzig , Germany
| |
Collapse
|
18
|
Marro BS, Blanc CA, Loring JF, Cahalan MD, Lane TE. Promoting remyelination: utilizing a viral model of demyelination to assess cell-based therapies. Expert Rev Neurother 2015; 14:1169-79. [PMID: 25245576 DOI: 10.1586/14737175.2014.955854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS. While a broad range of therapeutics effectively reduce the incidence of focal white matter inflammation and plaque formation for patients with relapse-remitting forms of MS, a challenge within the field is to develop therapies that allow for axonal protection and remyelination. In the last decade, growing interest has focused on utilizing neural precursor cells (NPCs) to promote remyelination. To understand how NPCs function in chronic demyelinating environments, several excellent pre-clinical mouse models have been developed. One well accepted model is infection of susceptible mice with neurotropic variants of mouse hepatitis virus (MHV) that undergo chronic demyelination exhibiting clinical and histopathologic similarities to MS patients. Combined with the possibility that an environmental agent such as a virus could trigger MS, the MHV model of demyelination presents a relevant mouse model to assess the therapeutic potential of NPCs transplanted into an environment in which inflammatory-mediated demyelination is established.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology and Biochemistry, University of California, Irvine 92697, USA
| | | | | | | | | |
Collapse
|
19
|
Weinger JG, Plaisted WC, Maciejewski SM, Lanier LL, Walsh CM, Lane TE. Activating receptor NKG2D targets RAE-1-expressing allogeneic neural precursor cells in a viral model of multiple sclerosis. Stem Cells 2015; 32:2690-701. [PMID: 24898518 DOI: 10.1002/stem.1760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 12/11/2022]
Abstract
Transplantation of major histocompatibility complex-mismatched mouse neural precursor cells (NPCs) into mice persistently infected with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in rapid rejection that is mediated, in part, by T cells. However, the contribution of the innate immune response to allograft rejection in a model of viral-induced neurological disease has not been well defined. Herein, we demonstrate that the natural killer (NK) cell-expressing-activating receptor NKG2D participates in transplanted allogeneic NPC rejection in mice persistently infected with JHMV. Cultured NPCs derived from C57BL/6 (H-2(b) ) mice express the NKG2D ligand retinoic acid early precursor transcript (RAE)-1 but expression was dramatically reduced upon differentiation into either glia or neurons. RAE-1(+) NPCs were susceptible to NK cell-mediated killing whereas RAE-1(-) cells were resistant to lysis. Transplantation of C57BL/6-derived NPCs into JHMV-infected BALB/c (H-2(d) ) mice resulted in infiltration of NKG2D(+) CD49b(+) NK cells and treatment with blocking antibody specific for NKG2D increased survival of allogeneic NPCs. Furthermore, transplantation of differentiated RAE-1(-) allogeneic NPCs into JHMV-infected BALB/c mice resulted in enhanced survival, highlighting a role for the NKG2D/RAE-1 signaling axis in allograft rejection. We also demonstrate that transplantation of allogeneic NPCs into JHMV-infected mice resulted in infection of the transplanted cells suggesting that these cells may be targets for infection. Viral infection of cultured cells increased RAE-1 expression, resulting in enhanced NK cell-mediated killing through NKG2D recognition. Collectively, these results show that in a viral-induced demyelination model, NK cells contribute to rejection of allogeneic NPCs through an NKG2D signaling pathway.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology & Biochemistry; Sue and Bill Gross Stem Cell Center, University of California, Irvine, California, USA
| | | | | | | | | | | |
Collapse
|
20
|
Weinger JG, Greenberg ML, Matheu MP, Parker I, Walsh CM, Lane TE, Cahalan MD. Two-photon imaging of cellular dynamics in the mouse spinal cord. J Vis Exp 2015. [PMID: 25742043 DOI: 10.3791/52580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Two-photon (2P) microscopy is utilized to reveal cellular dynamics and interactions deep within living, intact tissues. Here, we present a method for live-cell imaging in the murine spinal cord. This technique is uniquely suited to analyze neural precursor cell (NPC) dynamics following transplantation into spinal cords undergoing neuroinflammatory demyelinating disorders. NPCs migrate to sites of axonal damage, proliferate, differentiate into oligodendrocytes, and participate in direct remyelination. NPCs are thereby a promising therapeutic treatment to ameliorate chronic demyelinating diseases. Because transplanted NPCs migrate to the damaged areas on the ventral side of the spinal cord, traditional intravital 2P imaging is impossible, and only information on static interactions was previously available using histochemical staining approaches. Although this method was generated to image transplanted NPCs in the ventral spinal cord, it can be applied to numerous studies of transplanted and endogenous cells throughout the entire spinal cord. In this article, we demonstrate the preparation and imaging of a spinal cord with enhanced yellow fluorescent protein-expressing axons and enhanced green fluorescent protein-expressing transplanted NPCs.
Collapse
Affiliation(s)
- Jason G Weinger
- Molecular Biology and Biochemistry, University of California, Irvine
| | | | - Melanie P Matheu
- University of California San Francisco Diabetes Center, University of California, San Francisco
| | - Ian Parker
- Neurobiology and Behavior, University of California, Irvine
| | - Craig M Walsh
- Molecular Biology and Biochemistry, University of California, Irvine
| | | | | |
Collapse
|
21
|
Two-photon imaging of remyelination of spinal cord axons by engrafted neural precursor cells in a viral model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:E2349-55. [PMID: 24843159 DOI: 10.1073/pnas.1406658111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural precursor cells (NPCs) offer a promising approach for treating demyelinating diseases. However, the cellular dynamics that underlie transplanted NPC-mediated remyelination have not been described. Using two-photon imaging of a newly developed ventral spinal cord preparation and a viral model of demyelination, we describe the motility and intercellular interactions of transplanted mouse NPCs expressing green fluorescent protein (GFP) with damaged axons expressing yellow fluorescent protein (YFP). Our findings reveal focal axonal degeneration that occurs in the ventral side of the spinal cord within 1 wk following intracranial instillation with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Axonal damage precedes extensive demyelination and is characterized by swelling along the length of the axon, loss of YFP signal, and transected appearance. NPCs engrafted into spinal cords of JHMV-infected mice exhibited diminished migration velocities and increased proliferation compared with transplanted cells in noninfected mice. NPCs preferentially accumulated within areas of axonal damage, initiated direct contact with axons, and subsequently expressed the myelin proteolipid protein gene, initiating remyelination. These findings indicate that NPCs transplanted into an inflammatory demyelinating microenvironment participate directly in therapeutic outcome through the wrapping of myelin around damaged neurons.
Collapse
|
22
|
Plaisted WC, Weinger JG, Walsh CM, Lane TE. T cell mediated suppression of neurotropic coronavirus replication in neural precursor cells. Virology 2013; 449:235-43. [PMID: 24418558 PMCID: PMC3894587 DOI: 10.1016/j.virol.2013.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 09/17/2013] [Accepted: 11/14/2013] [Indexed: 02/02/2023]
Abstract
Neural precursor cells (NPCs) are the subject of intense investigation for their potential to treat neurodegenerative disorders, yet the consequences of neuroinvasive virus infection of NPCs remain unclear. This study demonstrates that NPCs support replication following infection by the neurotropic JHM strain of mouse hepatitis virus (JHMV). JHMV infection leads to increased cell death and dampens IFN-γ-induced MHC class II expression. Importantly, cytokines secreted by CD4+ T cells inhibit JHMV replication in NPCs, and CD8+ T cells specifically target viral peptide-pulsed NPCs for lysis. Furthermore, treatment with IFN-γ inhibits JHMV replication in a dose-dependent manner. Together, these findings suggest that T cells play a critical role in controlling replication of a neurotropic virus in NPCs, a finding which has important implications when considering immune modulation for NPC-based therapies for treatment of human neurologic diseases. Murine neural precursor cells are infected by JHMV in a CEACAM1a-dependent manner. Peptide-pulsed NPCs are targeted for lysis by virus-specific CD8+ T cells. JHMV replication in NPCs is suppressed by CD4+ T cells through IFN-γ secretion. IFN-γ dampens CEACAM1a expression and JHMV protein production in NPCs.
Collapse
Affiliation(s)
- Warren C Plaisted
- Department of Molecular Biology & Biochemistry, University of California, Irvine 92697-3900, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine 92697-3900, USA
| | - Jason G Weinger
- Department of Molecular Biology & Biochemistry, University of California, Irvine 92697-3900, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine 92697-3900, USA
| | - Craig M Walsh
- Department of Molecular Biology & Biochemistry, University of California, Irvine 92697-3900, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine 92697-3900, USA; Multiple Sclerosis Research Center, University of California, Irvine 92697-3900, USA; Institute for Immunology, University of California, Irvine 92697-3900, USA
| | - Thomas E Lane
- Department of Molecular Biology & Biochemistry, University of California, Irvine 92697-3900, USA; Sue and Bill Gross Stem Cell Center, University of California, Irvine 92697-3900, USA; Multiple Sclerosis Research Center, University of California, Irvine 92697-3900, USA; Institute for Immunology, University of California, Irvine 92697-3900, USA.
| |
Collapse
|
23
|
Weinger JG, Chen L, Coleman R, Leang R, Plaisted WC, Loring JF, Lane TE. Intraspinal transplantation of mouse and human neural precursor cells. ACTA ACUST UNITED AC 2013; 26:2D.16.1-2D.16.16. [PMID: 24510791 DOI: 10.1002/9780470151808.sc02d16s26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This unit describes the preparation and transplantation of human neural precursor cells (hNPCs) and mouse neural precursor cells (mNPCs) into the thoracic region of the mouse spinal cord. The techniques in this unit also describe how to prepare the mouse for surgery by performing a laminectomy to expose the spinal cord for transplantation. NPCs genetically labeled with eGFP transplanted into the spinal cord of a mouse following viral-mediated demyelination can efficiently be detected via eGFP expression. Transplantation of these cells into the spinal cord is an efficacious way to determine their effects in neurological disorders such as multiple sclerosis, Alzheimer's disease, and spinal cord injury.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, California
| | - Lu Chen
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, California
| | - Ronald Coleman
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California
| | - Ronika Leang
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, California
| | - Warren C Plaisted
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, California
| | - Jeanne F Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California
| | - Thomas E Lane
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, California
| |
Collapse
|
24
|
Pluchino S, Cossetti C. How stem cells speak with host immune cells in inflammatory brain diseases. Glia 2013; 61:1379-401. [PMID: 23633288 DOI: 10.1002/glia.22500] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/01/2013] [Indexed: 12/14/2022]
Abstract
Advances in stem cell biology have raised great expectations that diseases and injuries of the central nervous system (CNS) may be ameliorated by the development of non-hematopoietic stem cell medicines. Yet, the application of adult stem cells as CNS therapeutics is challenging and the interpretation of some of the outcomes ambiguous. In fact, the initial idea that stem cell transplants work only via structural cell replacement has been challenged by the observation of consistent cellular signaling between the graft and the host. Cellular signaling is the foundation of coordinated actions and flexible responses, and arises via networks of exchanging and interacting molecules that transmit patterns of information between cells. Sustained stem cell graft-to-host communication leads to remarkable trophic effects on endogenous brain cells and beneficial modulatory actions on innate and adaptive immune responses in vivo, ultimately promoting the healing of the injured CNS. Among a number of adult stem cell types, mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs) are being extensively investigated for their ability to signal to the immune system upon transplantation in experimental CNS diseases. Here, we focus on the main cellular signaling pathways that grafted MSCs and NPCs use to establish a therapeutically relevant cross talk with host immune cells, while examining the role of inflammation in regulating some of the bidirectionality of these communications. We propose that the identification of the players involved in stem cell signaling might contribute to the development of innovative, high clinical impact therapeutics for inflammatory CNS diseases.
Collapse
Affiliation(s)
- Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Cambridge Centre for Brain Repair and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, United Kingdom.
| | | |
Collapse
|