Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
EMBO Mol Med ; 15(9): e17748, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37538042

ABSTRACT

Hematopoietic cell transplantation (HCT) treats many blood conditions but remains underused due to complications such as graft-versus-host disease (GvHD). In GvHD, donor immune cells attack the patient, requiring powerful immunosuppressive drugs like glucocorticoids (GCs) to prevent death. In this study, we tested the hypothesis that donor cell conditioning with the glucocorticoid fluticasone propionate (FLU) prior to transplantation could increase hematopoietic stem cell (HSC) engraftment and reduce GvHD. Murine HSCs treated with FLU had increased HSC engraftment and reduced severity and incidence of GvHD after transplantation into allogeneic hosts. While most T cells died upon FLU treatment, donor T cells repopulated in the hosts and appeared less inflammatory and alloreactive. Regulatory T cells (Tregs) are immunomodulatory and survived FLU treatment, resulting in an increased ratio of Tregs to conventional T cells. Our results implicate an important role for Tregs in maintaining allogeneic tolerance in FLU-treated grafts and suggest a therapeutic strategy of pre-treating donor cells (and not the patients directly) with GCs to simultaneously enhance engraftment and reduce GvHD upon allogeneic HCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mice , Animals , Fluticasone/pharmacology , Fluticasone/therapeutic use , Transplantation, Homologous/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Graft vs Host Disease/prevention & control , Immunosuppressive Agents
2.
J Neuroimmunol ; 381: 578133, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37352687

ABSTRACT

Intracranial inoculation of susceptible mice with a glial-tropic strain of mouse hepatitis virus (JHMV), a murine coronavirus, results in an acute encephalomyelitis followed by viral persistence in white matter tracts accompanied by chronic neuroinflammation and demyelination. Microglia are the resident immune cell of the central nervous system (CNS) and are considered important in regulating events associated with neuroinflammation as well as influencing both white matter damage and remyelination. To better understand mechanisms by which microglia contribute to these immune-mediated events, JHMV-infected mice with established demyelination were treated with the small molecular inhibitor of colony stimulating factor 1 receptor (CSF1R), PLX5622, to deplete microglia. Treatment with PLX5622 did not affect viral replication within the CNS yet the severity of demyelination was increased and remyelination impaired compared to control mice. Gene expression analysis revealed that targeting microglia resulted in altered expression of genes associated with immune cell activation and phagocytosis of myelin debris. These findings indicate that microglia are not critical in viral surveillance in persistently JHMV-infected mice yet restrict white matter damage and remyelination, in part, by influencing phagocytosis of myelin debris.


Subject(s)
Coronavirus Infections , Demyelinating Diseases , Murine hepatitis virus , Remyelination , White Matter , Mice , Animals , Microglia/metabolism , Murine hepatitis virus/physiology , Neuroinflammatory Diseases , Coronavirus Infections/complications , Mice, Inbred C57BL
3.
J Immunol ; 210(11): 1677-1686, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37083696

ABSTRACT

Transplantation of human neural stem cells (hNSCs) is a promising regenerative therapy to promote remyelination in patients with multiple sclerosis (MS). Transplantation of hNSCs has been shown to increase the number of CD4+CD25+Foxp3+ T regulatory cells (Tregs) in the spinal cords of murine models of MS, which is correlated with a strong localized remyelination response. However, the mechanisms by which hNSC transplantation leads to an increase in Tregs in the CNS remains unclear. We report that hNSCs drive the conversion of T conventional (Tconv) cells into Tregs in vitro. Conversion of Tconv cells is Ag driven and fails to occur in the absence of TCR stimulation by cognate antigenic self-peptides. Furthermore, CNS Ags are sufficient to drive this conversion in the absence of hNSCs in vitro and in vivo. Importantly, only Ags presented in the thymus during T cell selection drive this Treg response. In this study, we investigate the mechanisms by which hNSC Ags drive the conversion of Tconv cells into Tregs and may provide key insight needed for the development of MS therapies.


Subject(s)
Multiple Sclerosis , Neural Stem Cells , Humans , Mice , Animals , T-Lymphocytes, Regulatory , CD4-Positive T-Lymphocytes , Multiple Sclerosis/therapy , Lymphocyte Activation , Forkhead Transcription Factors , CD4 Antigens
5.
Neurobiol Dis ; 140: 104868, 2020 07.
Article in English | MEDLINE | ID: mdl-32276110

ABSTRACT

Multiple sclerosis (MS) is a chronic, inflammatory autoimmune disease that affects the central nervous system (CNS) for which there is no cure. In MS, encephalitogenic T cells infiltrate the CNS causing demyelination and neuroinflammation; however, little is known about the role of regulatory T cells (Tregs) in CNS tissue repair. Transplantation of neural stem and progenitor cells (NSCs and NPCs) is a promising therapeutic strategy to promote repair through cell replacement, although recent findings suggest transplanted NSCs also instruct endogenous repair mechanisms. We have recently described that dampened neuroinflammation and increased remyelination is correlated with emergence of Tregs following human NPC transplantation in a murine viral model of immune-mediated demyelination. In the current study we utilized the prototypic murine autoimmune model of demyelination experimental autoimmune encephalomyelitis (EAE) to test the efficacy of hNSC transplantation. Eight-week-old, male EAE mice receiving an intraspinal transplant of hNSCs during the chronic phase of disease displayed remyelination, dampened neuroinflammation, and an increase in CNS CD4+CD25+FoxP3+ regulatory T cells (Tregs). Importantly, ablation of Tregs abrogated histopathological improvement. Tregs are essential for maintenance of T cell homeostasis and prevention of autoimmunity, and an emerging role for Tregs in maintenance of tissue homeostasis through interactions with stem and progenitor cells has recently been suggested. The data presented here provide direct evidence for collaboration between CNS Tregs and hNSCs promoting remyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis/therapy , Neural Stem Cells/transplantation , Remyelination , T-Lymphocytes, Regulatory , Animals , Humans , Male , Mice , Myelin Sheath , Stem Cell Transplantation
6.
Sci Immunol ; 5(44)2020 02 21.
Article in English | MEDLINE | ID: mdl-32086381

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses during cancer. It remains elusive how MDSCs differ from their normal myeloid counterparts, which limits our ability to specifically detect and therapeutically target MDSCs during cancer. Here, we sought to determine the molecular features of breast cancer-associated MDSCs using the widely studied mouse model based on the mouse mammary tumor virus (MMTV) promoter-driven expression of the polyomavirus middle T oncoprotein (MMTV-PyMT). To identify MDSCs in an unbiased manner, we used single-cell RNA sequencing to compare MDSC-containing splenic myeloid cells from breast tumor-bearing mice with wild-type controls. Our computational analysis of 14,646 single-cell transcriptomes revealed that MDSCs emerge through an aberrant neutrophil maturation trajectory in the spleen that confers them an immunosuppressive cell state. We establish the MDSC-specific gene signature and identify CD84 as a surface marker for improved detection and enrichment of MDSCs in breast cancers.


Subject(s)
Breast Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Single-Cell Analysis , Transcriptome , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Cell Differentiation/genetics , Female , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic , Myeloid-Derived Suppressor Cells/immunology , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology
7.
ACS Nano ; 13(6): 6670-6688, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31117376

ABSTRACT

To dissect therapeutic mechanisms of transplanted stem cells and develop exosome-based nanotherapeutics in treating autoimmune and neurodegenerative diseases, we assessed the effect of exosomes secreted from human mesenchymal stem cells (MSCs) in treating multiple sclerosis using an experimental autoimmune encephalomyelitis (EAE) mouse model. We found that intravenous administration of exosomes produced by MSCs stimulated by IFNγ (IFNγ-Exo) (i) reduced the mean clinical score of EAE mice compared to PBS control, (ii) reduced demyelination, (iii) decreased neuroinflammation, and (iv) upregulated the number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords of EAE mice. Co-culture of IFNγ-Exo with activated peripheral blood mononuclear cells (PBMCs) cells in vitro reduced PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines including IL-6, IL-12p70, IL-17AF, and IL-22 yet increased levels of immunosuppressive cytokine indoleamine 2,3-dioxygenase. IFNγ-Exo could also induce Tregs in vitro in a murine splenocyte culture, likely mediated by a third-party accessory cell type. Further, IFNγ-Exo characterization by deep RNA sequencing suggested that IFNγ-Exo contains anti-inflammatory RNAs, where their inactivation partially hindered the exosomes potential to induce Tregs. Furthermore, we found that IFNγ-Exo harbors multiple anti-inflammatory and neuroprotective proteins. These results not only shed light on stem cell therapeutic mechanisms but also provide evidence that MSC-derived exosomes can potentially serve as cell-free therapies in creating a tolerogenic immune response to treat autoimmune and central nervous system disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Exosomes/transplantation , Mesenchymal Stem Cell Transplantation/methods , Animals , Cells, Cultured , Exosomes/metabolism , Female , Humans , Interferon-gamma/pharmacology , Interleukins/genetics , Interleukins/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Monocytes/immunology , T-Lymphocytes, Regulatory/immunology
8.
Dev Dyn ; 248(1): 43-52, 2019 01.
Article in English | MEDLINE | ID: mdl-30067309

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.


Subject(s)
Neurodegenerative Diseases/therapy , Remyelination , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Humans , Mice , Multiple Sclerosis/therapy , Murine hepatitis virus , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/virology
9.
Glia ; 67(5): 844-856, 2019 05.
Article in English | MEDLINE | ID: mdl-30588668

ABSTRACT

Alzheimer's disease (AD) is the leading cause of age-related neurodegeneration and is characterized neuropathologically by the accumulation of insoluble beta-amyloid (Aß) peptides. In AD brains, plaque-associated myeloid (PAM) cells cluster around Aß plaques but fail to effectively clear Aß by phagocytosis. PAM cells were originally thought to be brain-resident microglia. However, several studies have also suggested that Aß-induced inflammation causes peripheral monocytes to enter the otherwise immune-privileged brain. The relationship between AD progression and inflammation in the brain remains ambiguous because microglia and monocyte-derived macrophages are extremely difficult to distinguish from one another in an inflamed brain. Whether PAM cells are microglia, peripheral macrophages, or a mixture of both remains unclear. CD11a is a component of the ß2 integrin LFA1. We have determined that CD11a is highly expressed on peripheral immune cells, including macrophages, but is not expressed by mouse microglia. These expression patterns remain consistent in LPS-treated inflamed mice, as well as in two mouse models of AD. Thus, CD11a can be used as a marker to distinguish murine microglia from infiltrating peripheral immune cells. Using CD11a, we show that PAM cells in AD transgenic brains are comprised entirely of microglia. We also demonstrate a novel fluorescence-assisted quantification technique (FAQT), which reveals a significant increase in T lymphocytes, especially in the brains of female AD mice. Our findings support the notion that microglia are the lead myeloid players in AD and that rejuvenating their phagocytic potential may be an important therapeutic strategy.


Subject(s)
Alzheimer Disease/pathology , CD11a Antigen/metabolism , Microglia/metabolism , Microglia/pathology , Myeloid Cells/metabolism , Algorithms , Alzheimer Disease/genetics , Alzheimer Disease/surgery , Animals , Animals, Newborn , Bone Marrow Transplantation , Brain/metabolism , Brain/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Humans , Inflammation/etiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Myeloid Cells/drug effects , Oxidoreductases/genetics , Oxidoreductases/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Toxoplasmosis/complications
10.
Virology ; 511: 49-55, 2017 11.
Article in English | MEDLINE | ID: mdl-28822268

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.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Murine hepatitis virus/growth & development , Murine hepatitis virus/immunology , Neural Stem Cells/immunology , Neural Stem Cells/virology , Animals , Carcinoembryonic Antigen/biosynthesis , Gene Expression , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Interferon-gamma/metabolism , Mice , Mice, Transgenic
11.
Proc Natl Acad Sci U S A ; 114(7): E1168-E1177, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137846

ABSTRACT

Disruption of the blood-brain barrier (BBB) is a defining and early feature of multiple sclerosis (MS) that directly damages the central nervous system (CNS), promotes immune cell infiltration, and influences clinical outcomes. There is an urgent need for new therapies to protect and restore BBB function, either by strengthening endothelial tight junctions or suppressing endothelial vesicular transcytosis. Although wingless integrated MMTV (Wnt)/ß-catenin signaling plays an essential role in BBB formation and maintenance in healthy CNS, its role in BBB repair in neurologic diseases such as MS remains unclear. Using a Wnt/ß-catenin reporter mouse and several downstream targets, we demonstrate that the Wnt/ß-catenin pathway is up-regulated in CNS endothelial cells in both human MS and the mouse model experimental autoimmune encephalomyelitis (EAE). Increased Wnt/ß-catenin activity in CNS blood vessels during EAE progression correlates with up-regulation of neuronal Wnt3 expression, as well as breakdown of endothelial cell junctions. Genetic inhibition of the Wnt/ß-catenin pathway in CNS endothelium before disease onset exacerbates the clinical presentation of EAE, CD4+ T-cell infiltration into the CNS, and demyelination by increasing expression of vascular cell adhesion molecule-1 and the transcytosis protein Caveolin-1 and promoting endothelial transcytosis. However, Wnt signaling attenuation does not affect the progressive degradation of tight junction proteins or paracellular BBB leakage. These results suggest that reactivation of Wnt/ß-catenin signaling in CNS vessels during EAE/MS partially restores functional BBB integrity and limits immune cell infiltration into the CNS.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Endothelial Cells/metabolism , Multiple Sclerosis/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Blood-Brain Barrier/metabolism , Caveolin 1/metabolism , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/genetics , Transcytosis , beta Catenin/genetics
13.
J Immunol ; 197(8): 3049-3058, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27613697

ABSTRACT

The mammalian target of rapamycin (mTOR) is essential for Th cell proliferation and effector differentiation, making the mTOR signaling network an attractive immunomodulatory target for autoimmune-related diseases. Although direct targeting of mTOR complex-1 (mTORC1) with rapamycin can provide clinical benefit, targeting downstream enzymes has the potential to offer more selective immunosuppression. In this study, we evaluated p70 ribosomal protein S6 Kinase 2 (S6K2), a downstream effector of mTORC1, for its role in T cell function and autoimmunity. S6K2 is a direct substrate of mTORC1, with a potential role in Th17 differentiation suggested by biochemical studies. Using a genetic approach with S6K2 knockout mice, we found that S6K2 loss reduces Th17 skewing and increases regulatory T cell differentiation in vitro when cultured in RPMI 1640 media. However, S6K2 was dispensable for Th17 differentiation in IMDM. In an in vivo experimental autoimmune encephalomyelitis model in which rapamycin suppresses disease, S6K2 knockout mice did not exhibit differences in clinical score or Th17 differentiation. These results suggest that S6K2 is dispensable for Th17-driven autoimmunity and highlight how distinct experimental conditions can produce significantly different results in T cell differentiation.


Subject(s)
Autoimmune Diseases/therapy , Immunologic Factors/therapeutic use , Multiprotein Complexes/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/immunology , Animals , Autoimmune Diseases/immunology , Autoimmunity , Cell Differentiation , Cells, Cultured , Immunosuppression Therapy , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Sirolimus/therapeutic use , Substrate Specificity
14.
PLoS One ; 11(6): e0157620, 2016.
Article in English | MEDLINE | ID: mdl-27310015

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.


Subject(s)
Coronavirus Infections/therapy , Embryoid Bodies/immunology , Hepatitis, Viral, Animal/therapy , Human Embryonic Stem Cells/immunology , Neural Stem Cells/transplantation , T-Lymphocytes, Regulatory/immunology , 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/virology , Disease Models, Animal , Embryoid Bodies/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Human Embryonic Stem Cells/cytology , 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 , Organ Specificity , T-Lymphocytes, Regulatory/pathology
15.
Biomaterials ; 77: 87-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26584349

ABSTRACT

Systemic administration of mesenchymal stem cells (MSCs) affords the potential to ameliorate the symptoms of Multiple Sclerosis (MS) in both preclinical and clinical studies. However, the efficacy of MSC-based therapy for MS likely depends on the number of cells that home to inflamed tissues and on the controlled production of paracrine and immunomodulatory factors. Previously, we reported that engineered MSCs expressing P-selectin glycoprotein ligand-1 (PSGL-1) and Sialyl-Lewis(x) (SLeX) via mRNA transfection facilitated the targeted delivery of anti-inflammatory cytokine interleukin-10 (IL-10) to inflamed ear. Here, we evaluated whether targeted delivery of MSCs with triple PSGL1/SLeX/IL-10 engineering improves therapeutic outcomes in mouse experimental autoimmune encephalomyelitis (EAE), a murine model for human MS. We found PSGL-1/SLeX mRNA transfection significantly enhanced MSC homing to the inflamed spinal cord. This is consistent with results from in vitro flow chamber assays in which PSGL-1/SleX mRNA transfection significantly increased the percentage of rolling and adherent cells on activated brain microvascular endothelial cells, which mimic the inflamed endothelium of blood brain/spinal cord barrier in EAE. In addition, IL-10-transfected MSCs show significant inhibitory activity on the proliferation of CD4(+) T lymphocytes from EAE mice. In vivo treatment with MSCs engineered with PSGL-1/SLeX/IL-10 in EAE mice exhibited a superior therapeutic function over native (unmodified) MSCs, evidenced by significantly improved myelination and decreased lymphocytes infiltration into the white matter of the spinal cord. Our strategy of targeted delivery of performance-enhanced MSCs could potentially be utilized to increase the effectiveness of MSC-based therapy for MS and other central nervous system (CNS) disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Interleukin-10/biosynthesis , Membrane Glycoproteins/biosynthesis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion , Cell Movement , Coculture Techniques , Endothelium, Vascular/metabolism , Genetic Vectors/genetics , HL-60 Cells , Humans , Interleukin-10/genetics , Lentivirus/genetics , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Myelin Sheath/physiology , N-Acetylneuraminic Acid/metabolism , Organ Specificity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Spinal Cord/pathology , Transfection
17.
J Vis Exp ; (96)2015 Feb 22.
Article in English | MEDLINE | ID: mdl-25742043

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.


Subject(s)
Microscopy, Fluorescence, Multiphoton/methods , Neural Stem Cells/cytology , Spinal Cord/cytology , Animals , Axons/physiology , Cell Differentiation/physiology , Cells, Cultured , Demyelinating Diseases , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Neurons/cytology , Oligodendroglia/cytology
18.
Int Rev Immunol ; 34(1): 3-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398050

ABSTRACT

While simultaneously maintaining homeostasis and reducing further harm to the host, the immune system is equipped to eliminate both tumors and pathogenic microorganisms. Bifurcated into cell-mediated and humoral immunity, the adaptive immune system requires a series of complex and coordinated signals to drive the proliferation and differentiation of appropriate subsets. These include signals that modulate cellular metabolism. When first published in the 1920s, "the Warburg effect" was used to describe a phenomenon in which most cancer cells relied on aerobic glycolysis to meet their biosynthetic demands. Despite the early observations of Warburg and his colleagues, targeting cancer cell metabolism for therapeutic purposes still remains theoretical. Notably, many T cells exhibit the same Warburg metabolism as cancer cells and the therapeutic benefit of targeting their metabolic pathways has since been reexamined. Emerging evidence suggests that specific metabolic alterations associated with T cells may be ancillary to their subset differentiation and influential in their inflammatory response. Thus, T cell lymphocyte activation leads to skewing in metabolic plasticity, and issue that will be the subject of this review.


Subject(s)
Glucose/metabolism , Glycolysis/immunology , Immunity, Cellular , Immunity, Humoral , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Glucose/immunology , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/immunology , Glycolysis/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Leptin/genetics , Leptin/immunology , Lymphocyte Activation , Oxidative Phosphorylation , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
19.
J Leukoc Biol ; 97(1): 147-60, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25381385

ABSTRACT

A complete genetic deficiency of the complement protein C1q results in SLE with nearly 100% penetrance in humans, but the molecular mechanisms responsible for this association have not yet been fully determined. C1q opsonizes ACs for enhanced ingestion by phagocytes, such as Mϕ and iDCs, avoiding the extracellular release of inflammatory DAMPs upon loss of the membrane integrity of the dying cell. We previously showed that human monocyte-derived Mϕ and DCs ingesting autologous, C1q-bound LALs (C1q-polarized Mϕ and C1q-polarized DCs), enhance the production of anti-inflammatory cytokines, and reduce proinflammatory cytokines relative to Mϕ or DC ingesting LAL alone. Here, we show that C1q-polarized Mϕ have elevated PD-L1 and PD-L2 and suppressed surface CD40, and C1q-polarized DCs have higher surface PD-L2 and less CD86 relative to Mϕ or DC ingesting LAL alone, respectively. In an MLR, C1q-polarized Mϕ reduced allogeneic and autologous Th17 and Th1 subset proliferation and demonstrated a trend toward increased Treg proliferation relative to Mϕ ingesting LAL alone. Moreover, relative to DC ingesting AC in the absence of C1q, C1q-polarized DCs decreased autologous Th17 and Th1 proliferation. These data demonstrate that a functional consequence of C1q-polarized Mϕ and DC is the regulation of Teff activation, thereby "sculpting" the adaptive immune system to avoid autoimmunity, while clearing dying cells. It is noteworthy that these studies identify novel target pathways for therapeutic intervention in SLE and other autoimmune diseases.


Subject(s)
Cell Proliferation , Complement C1q/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Apoptosis/immunology , Flow Cytometry , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Culture Test, Mixed , Phagocytosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...