Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 92020 12 02.
Article in English | MEDLINE | ID: mdl-33263277

ABSTRACT

Sciatic nerve crush injury triggers sterile inflammation within the distal nerve and axotomized dorsal root ganglia (DRGs). Granulocytes and pro-inflammatory Ly6Chigh monocytes infiltrate the nerve first and rapidly give way to Ly6Cnegative inflammation-resolving macrophages. In axotomized DRGs, few hematogenous leukocytes are detected and resident macrophages acquire a ramified morphology. Single-cell RNA-sequencing of injured sciatic nerve identifies five macrophage subpopulations, repair Schwann cells, and mesenchymal precursor cells. Macrophages at the nerve crush site are molecularly distinct from macrophages associated with Wallerian degeneration. In the injured nerve, macrophages 'eat' apoptotic leukocytes, a process called efferocytosis, and thereby promote an anti-inflammatory milieu. Myeloid cells in the injured nerve, but not axotomized DRGs, strongly express receptors for the cytokine GM-CSF. In GM-CSF-deficient (Csf2-/-) mice, inflammation resolution is delayed and conditioning-lesion-induced regeneration of DRG neuron central axons is abolished. Thus, carefully orchestrated inflammation resolution in the nerve is required for conditioning-lesion-induced neurorepair.


Subject(s)
Ganglia, Spinal/immunology , Leukocytes/immunology , Macrophages/immunology , Nerve Regeneration , Peripheral Nerve Injuries/immunology , Phagocytosis , Sciatic Nerve/immunology , Animals , Apoptosis , Cells, Cultured , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation , Gene Regulatory Networks , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation Mediators/metabolism , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Neuronal Outgrowth , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction
2.
Nat Immunol ; 21(12): 1496-1505, 2020 12.
Article in English | MEDLINE | ID: mdl-33106668

ABSTRACT

Transected axons typically fail to regenerate in the central nervous system (CNS), resulting in chronic neurological disability in individuals with traumatic brain or spinal cord injury, glaucoma and ischemia-reperfusion injury of the eye. Although neuroinflammation is often depicted as detrimental, there is growing evidence that alternatively activated, reparative leukocyte subsets and their products can be deployed to improve neurological outcomes. In the current study, we identify a unique granulocyte subset, with characteristics of an immature neutrophil, that had neuroprotective properties and drove CNS axon regeneration in vivo, in part via secretion of a cocktail of growth factors. This pro-regenerative neutrophil promoted repair in the optic nerve and spinal cord, demonstrating its relevance across CNS compartments and neuronal populations. Our findings could ultimately lead to the development of new immunotherapies that reverse CNS damage and restore lost neurological function across a spectrum of diseases.


Subject(s)
Axons/metabolism , Cell Communication , Central Nervous System/cytology , Central Nervous System/metabolism , Nerve Regeneration , Neurons/metabolism , Neutrophils/metabolism , Animals , Biomarkers , Cell Plasticity/immunology , Cell Survival/drug effects , Cell Survival/immunology , Central Nervous System/immunology , Intercellular Signaling Peptides and Proteins/biosynthesis , Mice , Neutrophil Infiltration/immunology , Neutrophils/immunology , Optic Nerve/immunology , Optic Nerve/metabolism , Receptors, Interleukin-8B/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Transcriptome , Zymosan/metabolism , Zymosan/pharmacology
3.
J Immunol ; 195(6): 2552-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26238492

ABSTRACT

Multiple sclerosis (MS) is believed to be initiated by myelin-reactive CD4(+) Th cells. IL-12-polarized Th1 cells, IL-23-polarized Th17 cells, and Th17 cells that acquire Th1 characteristics were each implicated in autoimmune pathogenesis. It is debated whether Th cells that can drive the development of demyelinating lesions are phenotypically diverse or arise from a single lineage. In the current study, we assessed the requirement of IL-12 or IL-23 stimulation, as well as Th plasticity, for the differentiation of T cells capable of inducing CNS axon damage. We found that stable murine Th1 and Th17 cells independently transfer experimental autoimmune encephalomyelitis (widely used as an animal model of MS) in the absence of IL-23 and IL-12, respectively. Plastic Th17 cells are particularly potent mediators of demyelination and axonopathy. In parallel studies, we identified MS patients who consistently mount either IFN-γ- or IL-17-skewed responses to myelin basic protein over the course of a year. Brain magnetic resonance imaging revealed that patients with mixed IFN-γ and IL-17 responses have relatively high T1 lesion burden, a measure of permanent axon damage. Our data challenge the dogma that IL-23 and Th17 plasticity are universally required for the development of experimental autoimmune encephalomyelitis. This study definitively demonstrates that autoimmune demyelinating disease can be driven by distinct Th-polarizing factors and effector subsets, underscoring the importance of a customized approach to the pharmaceutical management of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Autoimmunity/immunology , Brain/diagnostic imaging , Cell Differentiation/immunology , Demyelinating Diseases/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/immunology , Optic Nerve/immunology , Optic Nerve/pathology , Radiography , Th1 Cells/cytology , Th1 Cells/transplantation , Th17 Cells/cytology , Th17 Cells/transplantation
4.
Proc Natl Acad Sci U S A ; 112(8): 2581-6, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675510

ABSTRACT

Innate immunity can facilitate nervous system regeneration, yet the underlying cellular and molecular mechanisms are not well understood. Here we show that intraocular injection of lipopolysaccharide (LPS), a bacterial cell wall component, or the fungal cell wall extract zymosan both lead to rapid and comparable intravitreal accumulation of blood-derived myeloid cells. However, when combined with retro-orbital optic nerve crush injury, lengthy growth of severed retinal ganglion cell (RGC) axons occurs only in zymosan-injected mice, and not in LPS-injected mice. In mice deficient for the pattern recognition receptor dectin-1 but not Toll-like receptor-2 (TLR2), zymosan-mediated RGC regeneration is greatly reduced. The combined loss of dectin-1 and TLR2 completely blocks the proregenerative effects of zymosan. In the retina, dectin-1 is expressed by microglia and dendritic cells, but not by RGCs. Dectin-1 is also present on blood-derived myeloid cells that accumulate in the vitreous. Intraocular injection of the dectin-1 ligand curdlan [a particulate form of ß(1, 3)-glucan] promotes optic nerve regeneration comparable to zymosan in WT mice, but not in dectin-1(-/-) mice. Particulate ß(1, 3)-glucan leads to increased Erk1/2 MAP-kinase signaling and cAMP response element-binding protein (CREB) activation in myeloid cells in vivo. Loss of the dectin-1 downstream effector caspase recruitment domain 9 (CARD9) blocks CREB activation and attenuates the axon-regenerative effects of ß(1, 3)-glucan. Studies with dectin-1(-/-)/WT reciprocal bone marrow chimeric mice revealed a requirement for dectin-1 in both retina-resident immune cells and bone marrow-derived cells for ß(1, 3)-glucan-elicited optic nerve regeneration. Collectively, these studies identify a molecular framework of how innate immunity enables repair of injured central nervous system neurons.


Subject(s)
Axons/physiology , Central Nervous System/pathology , Inflammation/pathology , Lectins, C-Type/metabolism , Nerve Regeneration/drug effects , Signal Transduction/drug effects , beta-Glucans/adverse effects , Animals , CARD Signaling Adaptor Proteins/metabolism , Central Nervous System/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Phagocytosis/drug effects , Radiation Tolerance/drug effects , Retina/drug effects , Retina/metabolism , Toll-Like Receptor 2/metabolism , Zymosan/pharmacology
5.
Proc Natl Acad Sci U S A ; 111(22): E2349-55, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843159

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.


Subject(s)
Axons/physiology , Multiple Sclerosis/therapy , Myelin Sheath/physiology , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Stem Cell Transplantation/methods , Animals , Bacterial Proteins/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Demyelinating Diseases/therapy , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Hepatitis, Viral, Animal/complications , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Multiple Sclerosis/virology , Murine hepatitis virus , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spinal Cord/cytology
6.
Glia ; 59(12): 1813-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21830237

ABSTRACT

Following intracranial infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV), susceptible mice will develop widespread myelin destruction that results in pathological and clinical outcomes similar to those seen in humans with the demyelinating disease Multiple Sclerosis (MS). Partial remyelination and clinical recovery occurs during the chronic phase following control of viral replication yet the signaling mechanisms regulating these events remain enigmatic. Here we report the kinetics of proliferation and maturation of oligodendrocyte progenitor cells (OPCs) within the spinal cord following JHMV-induced demyelination and that CXCR4 signaling contributes to the maturation state of OPCs. Following treatment with AMD3100, a specific inhibitor of CXCR4, mice recovering from widespread demyelination exhibit a significant (P < 0.01) increase in the number of OPCs and fewer (P < 0.05) mature oligodendrocytes compared with HBSS-treated animals. These results suggest that CXCR4 signaling is required for OPCs to mature and contribute to remyelination in response to JHMV-induced demyelination. To assess if this effect is reversible and has potential therapeutic benefit, we pulsed mice with AMD3100 and then allowed them to recover. This treatment strategy resulted in increased numbers of mature oligodendrocytes, enhanced remyelination, and improved clinical outcome. These findings highlight the possibility to manipulate OPCs in order to increase the pool of remyelination-competent cells that can participate in recovery.


Subject(s)
Demyelinating Diseases/metabolism , Myelin Sheath/pathology , Myelin Sheath/virology , Oligodendroglia/pathology , Receptors, CXCR4/physiology , Signal Transduction/physiology , Stem Cells/pathology , Animals , Demyelinating Diseases/physiopathology , Demyelinating Diseases/virology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Oligodendroglia/virology , Primary Cell Culture , Receptors, CXCR4/antagonists & inhibitors , Stem Cells/virology
7.
J Vis Exp ; (53): e2834, 2011 Jul 10.
Article in English | MEDLINE | ID: mdl-21775959

ABSTRACT

Mice infected with the neurotropic JHM strain of mouse hepatitis virus (MHV) develop pathological and clinical outcomes similar to patients with the demyelinating disease Multiple Sclerosis (MS). We have shown that transplantation of NSCs into the spinal cords of sick mice results in a significant improvement in both remyelination and in clinical outcome. Cell replacement therapies for the treatment of chronic neurologic diseases are now a reality and in vivo models are vital in understanding the interactions between the engrafted cells and host tissue microenvironment. This presentation provides an adapted method for transplanting cells into the spinal cord of JHMV-infected mice. In brief, we provide a procedure for i) preparation of NSCs prior to transplant, ii) pre-operative care of mice, iii) exposure of the spinal cord via laminectomy, iv) stereotactic injection of NSCs, and iv) post-operative care.


Subject(s)
Demyelinating Diseases/surgery , Demyelinating Diseases/virology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/surgery , Neural Stem Cells/transplantation , Spinal Cord/cytology , Stem Cell Transplantation/methods , Animals , Laminectomy/methods , Mice , Murine hepatitis virus , Spinal Cord/pathology , Spinal Cord/virology
8.
J Neuroimmunol ; 224(1-2): 101-7, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20627412

ABSTRACT

Persistent infection of the central nervous system (CNS) of mice with the neuroadapted JHM strain of mouse hepatitis (MHV) is characterized by ongoing demyelination mediated by inflammatory T cells and macrophages that is similar both clinically and histologically with the human demyelinating disease multiple sclerosis (MS). Although extensive demyelination occurs in mice persistently infected with MHV there is only limited remyelination. Therefore, the MHV model of demyelination is a relevant model for studying disease and evaluating therapeutic approaches to protect cells of the oligodendrocyte lineage and promote remyelination. This concept is further highlighted as the etiology of MS remains enigmatic, but viruses have long been considered as potential triggering agents in initiating and/or maintaining MS symptoms. As such, understanding mechanisms associated with promoting repair within the CNS in the context of a persistent viral infection is critical given the possible viral etiology of MS. This review focuses on recent studies using either mouse neural stem cells (NSCs) or human oligodendrocyte progenitor cells (OPCs) derived from human embryonic stem cell (hESC) to promote remyelination in mice persistently infected with MHV. In addition, the potential role for chemokines in positional migration of transplanted cells is addressed.


Subject(s)
Coronavirus Infections/therapy , Demyelinating Diseases/therapy , Demyelinating Diseases/virology , Encephalitis, Viral/therapy , Murine hepatitis virus/immunology , Nerve Regeneration/immunology , Stem Cell Transplantation/methods , Animals , Cell Lineage/immunology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/pathology , Humans , Mice , Nerve Fibers, Myelinated/immunology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Stem Cell Transplantation/trends
9.
Proc Natl Acad Sci U S A ; 107(24): 11068-73, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20534452

ABSTRACT

Multiple sclerosis (MS) is a human demyelinating disease characterized by multifocal regions of inflammation, progressive myelin loss within the central nervous system (CNS), and eventual failure to remyelinate damaged axons. These problems suggest deficiencies in recruiting and/or maturation of oligodendrocyte progentior cells (OPCs) and highlight cell replacement therapies to promote remyelination. We have used a model of viral-induced demyelination to characterize signaling cues associated with positional migration of transplanted remyelination-competent cells. Although successful transplantation of rodent-derived glial cell types into models of MS has been performed, the mechanisms by which these cells navigate within an inflammatory environment created by a persistent virus has not been defined. Infection of the mouse CNS with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in an immune-mediated demyelinating disease with clinical and histologic similarities to MS. Surgical engraftment of GFP+ neural stem cells (NSCs) into spinal cords of JHMV-infected mice with established demyelination results in migration, proliferation, and differentiation of the cells into OPCs and mature oligodendrocytes that is associated with increased axonal remyelination. Treatment with anti-CXCL12 [stromal derived factor-1alpha, (SDF-1alpha)] blocking serum resulted in a marked impairment in migration and proliferation of engrafted stem cells. Moreover, small molecule-mediated antagonism of CXCR4, but not CXCR7, impaired migration and proliferation, to an extent similar to that with anti-CXCL12 treatment. These data highlight the importance of the CXCL12:CXCR4 pathway in regulating homing of engrafted stem cells to sites of tissue damage within the CNS of mice persistently infected with a neurotropic virus undergoing immune-mediated demyelination.


Subject(s)
Adult Stem Cells/physiology , Adult Stem Cells/transplantation , Chemokine CXCL12/physiology , Multiple Sclerosis/physiopathology , Neurons/physiology , Neurons/transplantation , Receptors, CXCR4/physiology , Animals , Cell Movement/physiology , Cell Proliferation , Disease Models, Animal , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Murine hepatitis virus/pathogenicity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...