Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Neurobiol Dis ; 190: 106370, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38049013

ABSTRACT

After spinal cord injury (SCI), infiltrating macrophages undergo excessive phagocytosis of myelin and cellular debris, forming lipid-laden foamy macrophages. To understand their role in the cellular pathology of SCI, investigation of the foamy macrophage phenotype in vitro revealed a pro-inflammatory profile, increased reactive oxygen species (ROS) production, and mitochondrial dysfunction. Bioinformatic analysis identified PI3K as a regulator of inflammation in foamy macrophages, and inhibition of this pathway decreased their lipid content, inflammatory cytokines, and ROS production. Macrophage-specific inhibition of PI3K using liposomes significantly decreased foamy macrophages at the injury site after a mid-thoracic contusive SCI in mice. RNA sequencing and in vitro analysis of foamy macrophages revealed increased autophagy and decreased phagocytosis after PI3K inhibition as potential mechanisms for reduced lipid accumulation. Together, our data suggest that the formation of pro-inflammatory foamy macrophages after SCI is due to the activation of PI3K signaling, which increases phagocytosis and decreases autophagy.


Subject(s)
Phosphatidylinositol 3-Kinases , Spinal Cord Injuries , Mice , Animals , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Macrophages/metabolism , Spinal Cord Injuries/metabolism , Lipids , Spinal Cord/pathology
2.
Br J Haematol ; 201(6): 1129-1143, 2023 06.
Article in English | MEDLINE | ID: mdl-36990798

ABSTRACT

Oral azacitidine (Oral-AZA) maintenance therapy improved relapse-free (RFS) and overall survival (OS) significantly versus placebo for AML patients in remission after intensive chemotherapy (IC) in the phase 3 QUAZAR AML-001 study. Immune profiling was performed on the bone marrow (BM) at remission and on-treatment in a subset of patients with the aim of identifying prognostic immune features and evaluating associations of on-treatment immune effects by Oral-AZA with clinical outcomes. Post-IC, increased levels of lymphocytes, monocytes, T cells and CD34 + CD117+ BM cells were prognostically favourable for RFS. CD3+ T-cell counts were significantly prognostic for RFS in both treatment arms. At baseline, high expression of the PD-L1 checkpoint marker was identified on a subset of CD34 + CD117+ BM cells; many of which were PD-L2+. High co-expression of T-cell exhaustion markers PD-1 and TIM-3 was associated with inferior outcomes. Oral-AZA augmented T-cell numbers during early treatment, increased CD4+:CD8+ ratios and reversed T-cell exhaustion. Unsupervised clustering analysis identified two patient subsets defined by T-cell content and expression of T-cell exhaustion markers that were enriched for MRD negativity. These results indicate that Oral-AZA modulates T-cell activity in the maintenance setting of AML, and these immune-mediated responses are associated with clinical outcomes.


Subject(s)
Bone Marrow , Leukemia, Myeloid, Acute , Humans , Neoplasm Recurrence, Local/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites/therapeutic use , Antigens, CD34 , Azacitidine/pharmacology , Azacitidine/therapeutic use , Tumor Microenvironment
3.
Hear Res ; 428: 108680, 2023 02.
Article in English | MEDLINE | ID: mdl-36586170

ABSTRACT

OBJECTIVE: Mild therapeutic hypothermia (MTH) has been demonstrated to prevent residual hearing loss from surgical trauma associated with cochlear implant (CI) insertion. Here, we aimed to characterize the mechanisms of MTH-induced hearing preservation in CI in a well-established preclinical rodent model. APPROACH: Rats were divided into four experimental conditions: MTH-treated and implanted cochleae, cochleae implanted under normothermic conditions, MTH only cochleae and un-operated cochleae (controls). Auditory brainstem responses (ABRs) were recorded at different time points (up to 84 days) to confirm long-term protection and safety of MTH locally applied to the cochlea for 20 min before and after implantation. Transcriptome sequencing profiling was performed on cochleae harvested 24 h post CI and MTH treatment to investigate the potential beneficial effects and underlying active gene expression pathways targeted by the temperature management. RESULTS: MTH treatment preserved residual hearing up to 3 months following CI when compared to the normothermic CI group. In addition, MTH applied locally to the cochleae using our surgical approach was safe and did not affect hearing in the long-term. Results of RNA sequencing analysis highlight positive modulation of signaling pathways and gene expression associated with an activation of cellular inflammatory and immune responses against the mechanical damage caused by electrode insertion. SIGNIFICANCE: These data suggest that multiple and possibly independent molecular pathways play a role in the protection of residual hearing provided by MTH against the trauma of cochlear implantation.


Subject(s)
Cochlear Implantation , Cochlear Implants , Hearing Loss , Hypothermia, Induced , Rats , Animals , Cochlear Implantation/adverse effects , Cochlear Implants/adverse effects , Cochlea/injuries , Hearing Loss/genetics , Hearing Loss/prevention & control , Hypothermia, Induced/methods
4.
Mol Neurobiol ; 59(12): 7393-7403, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36181661

ABSTRACT

Although glial scar formation has been extensively studied after optic nerve injury, the existence and characteristics of traumatic optic nerve fibrotic scar formation have not been previously characterized. Recent evidence suggests infiltrating macrophages are involved in pathological processes after optic nerve crush (ONC), but their role in fibrotic scar formation is unknown. Using wild-type and transgenic mouse models with optic nerve crush injury, we show that macrophages infiltrate and associate with fibroblasts in the traumatic optic nerve lesion fibrotic scar. We dissected the role of hematogenous and resident macrophages, labeled with Dil liposomes intravenously administered, and observed that hematogenous macrophages (Dil+ cells) specifically accumulate in the center of traumatic fibrotic scar while Iba-1+ cells reside predominantly at the margins of optic nerve fibrotic scar. Depletion of hematogenous macrophages results in reduced fibroblast density and decreased extracellular matrix deposition within the fibrotic scar area following ONC. However, retinal ganglion cell degeneration and function loss after optic nerve crush remain unaffected after hematogenous macrophage depletion. We present new and previously not characterized evidence that hematogenous macrophages are selectively recruited into the fibrotic core of the optic nerve crush site and critical for this fibrotic scar formation.


Subject(s)
Cicatrix , Optic Nerve Injuries , Mice , Animals , Cicatrix/pathology , Nerve Regeneration/physiology , Nerve Crush , Optic Nerve/pathology , Optic Nerve Injuries/pathology , Macrophages/pathology , Mice, Transgenic , Fibrosis , Disease Models, Animal
5.
Brief Funct Genomics ; 21(5): 339-356, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35822343

ABSTRACT

Cell-cell interactions (CCIs) are essential for multicellular organisms to coordinate biological processes and functions. One classical type of CCI interaction is between secreted ligands and cell surface receptors, i.e. ligand-receptor (LR) interactions. With the recent development of single-cell technologies, a large amount of single-cell ribonucleic acid (RNA) sequencing (scRNA-Seq) data has become widely available. This data availability motivated the single-cell-resolution study of CCIs, particularly LR-based CCIs. Dozens of computational methods and tools have been developed to predict CCIs by identifying LR-based CCIs. Many of these tools have been theoretically reviewed. However, there is little study on current LR-based CCI prediction tools regarding their performance and running results on public scRNA-Seq datasets. In this work, to fill this gap, we tested and compared nine of the most recent computational tools for LR-based CCI prediction. We used 15 well-studied scRNA-Seq samples that correspond to approximately 100K single cells under different experimental conditions for testing and comparison. Besides briefing the methodology used in these nine tools, we summarized the similarities and differences of these tools in terms of both LR prediction and CCI inference between cell types. We provided insight into using these tools to make meaningful discoveries in understanding cell communications.


Subject(s)
Single-Cell Analysis , Software , Cell Communication , Gene Expression Profiling/methods , Ligands , RNA , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
6.
Exp Neurol ; 355: 114147, 2022 09.
Article in English | MEDLINE | ID: mdl-35738417

ABSTRACT

Following injury in the central nervous system, a population of astrocytes occupy the lesion site, form glial bridges and facilitate axon regeneration. These astrocytes originate primarily from resident astrocytes or NG2+ oligodendrocyte progenitor cells. However, the extent to which these cell types give rise to the lesion-filling astrocytes, and whether the astrocytes derived from different cell types contribute similarly to optic nerve regeneration remain unclear. Here we examine the distribution of astrocytes and NG2+ cells in an optic nerve crush model. We show that optic nerve astrocytes partially fill the injury site over time after a crush injury. Viral mediated expression of a growth-promoting factor, ciliary neurotrophic factor (CNTF), in retinal ganglion cells (RGCs) promotes axon regeneration without altering the lesion size or the degree of lesion-filling GFAP+ cells. Strikingly, using inducible NG2CreER driver mice, we found that CNTF overexpression in RGCs increases the occupancy of NG2+ cell-derived astrocytes in the optic nerve lesion. An EdU pulse-chase experiment shows that the increase in NG2 cell-derived astrocytes is not due to an increase in cell proliferation. Lastly, we performed RNA-sequencing on the injured optic nerve and reveal that CNTF overexpression in RGCs results in significant changes in the expression of distinct genes, including those that encode chemokines, growth factor receptors, and immune cell modulators. Even though CNTF-induced axon regeneration has long been recognized, this is the first evidence of this procedure affecting glial cell fate at the optic nerve crush site. We discuss possible implication of these results for axon regeneration.


Subject(s)
Optic Nerve Injuries , Trauma, Nervous System , Animals , Astrocytes/metabolism , Axons/pathology , Ciliary Neurotrophic Factor , Cytokines/metabolism , Mice , Nerve Regeneration/physiology , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/metabolism , Trauma, Nervous System/metabolism
7.
Exp Neurol ; 352: 114035, 2022 06.
Article in English | MEDLINE | ID: mdl-35276206

ABSTRACT

Spinal cord injuries (SCI) often lead to multiple neurological deficits as a result from the initial trauma and also the secondary damage that follows. Despite abundant preclinical data proposing anti-inflammatory therapies to minimize secondary injury and improve functional recovery, the field still lacks an effective neuroprotective treatment. Epigenetic proteins, such as bromodomain and extraterminal domain (BET) proteins, are emerging as new targets to regulate inflammation. More importantly, pharmacological inhibition of BET proteins suppresses pro-inflammatory gene transcription after SCI. In this study, we tested the therapeutic potential of inhibiting BET proteins after SCI with clinically relevant compounds, and investigated the role of the BET protein BRD4 in macrophages during progression of SCI pathology. Systemic inhibition of BET proteins with I-BET762 significantly reduced lesion size 8 weeks after a contusion injury in rats. However, we observed no histological or locomotor improvements after SCI when we deleted Brd4 in macrophages through the use of myeloid-specific Brd4 knockout mice or after macrophage-targeted pharmacological BET inhibition. Taken together, our data indicate that systemic I-BET762 treatment is neuroprotective, and the histopathological improvement observed is likely to be a result of effects on non-macrophage targets. Expanding our understanding on the role of BET proteins after SCI is necessary to identify novel therapeutic targets that can effectively promote repair after SCI.


Subject(s)
Neuroprotection , Spinal Cord Injuries , Animals , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Rats , Recovery of Function/physiology , Rodentia , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Neurobiol Dis ; 163: 105608, 2022 02.
Article in English | MEDLINE | ID: mdl-34979258

ABSTRACT

Tissue damage after spinal cord injury (SCI) elicits a robust inflammatory cascade that fails to resolve in a timely manner, resulting in impaired wound healing and cellular regeneration. This inflammatory response is partly mediated by infiltrating immune cells, including macrophages. As professional phagocytes, macrophages initially play an important role in debris clearance at the injury site, which would be necessary for proper tissue regeneration. After SCI, most macrophages become filled with lipid droplets due to excessive uptake of lipid debris, assuming a "foamy" phenotype that is associated with a proinflammatory state. Myelin has been assumed to be the main source of lipid that induces foamy macrophage formation after injury given its abundance in the spinal cord. This assumption has led to the widespread use of purified myelin treatment to model foamy macrophage formation in vitro. However, the assumption that myelin is necessary for foamy macrophage formation remains untested. To this end, we developed a novel foamy macrophage assay utilizing total spinal cord homogenate to include all sources of lipid present at the injury site. Using the myelin basic protein knockout (MBP KO, i.e., Shiverer) mice that lack myelin, we investigated lipid accumulation in foamy macrophages. Primary macrophages treated with myelin-deficient spinal cord homogenate still formed large lipid droplets typically observed in foamy macrophages, although to a lesser degree than cells treated with normal homogenate. Similarly, MBP KO mice subjected to contusive spinal cord injury also formed foamy macrophages that exhibited reduced lipid content and associated with improved histological outcomes and reduced immune cell infiltration. Therefore, the absence of myelin does not preclude foamy macrophage formation, indicating that myelin is not the only major source of lipid that contributes this pathology, even though myelin may alter certain aspects of its inflammatory profile.


Subject(s)
Macrophages/pathology , Myelin Sheath/pathology , Spinal Cord Injuries/pathology , Spinal Cord/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/metabolism , Inflammation/pathology , Lipids , Macrophage Activation/physiology , Macrophages/metabolism , Male , Mice , Myelin Sheath/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
9.
Cell Tissue Res ; 387(3): 351-360, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34189605

ABSTRACT

Formation of a collagenous connective tissue scar after penetrating injuries to the brain or spinal cord has been described and investigated for well over 100 years. However, it was studied almost exclusively in the context of penetrating injuries that resulted in infiltration of meningeal fibroblasts, which raised doubts about translational applicability to most CNS injuries where the meninges remain intact. Recent studies demonstrating the perivascular niche as a source of fibroblasts have debunked the traditional view that a fibrotic scar only forms after penetrating lesions that tear the meninges. These studies have led to a renewed interest in CNS fibrosis not only in the context of axon regeneration after spinal cord injury, but also across a spectrum of CNS disorders. Arising with this renewed interest is some discrepancy about which perivascular cell gives rise to the fibrotic scar, but additional studies are beginning to provide some clarity. Although mechanistic studies on CNS fibrosis are still lacking, the similarities to fibrosis of other organs should provide important insight into how CNS fibrosis can be therapeutically targeted to promote functional recovery.


Subject(s)
Axons , Spinal Cord Injuries , Astrocytes/pathology , Central Nervous System , Cicatrix/pathology , Fibrosis , Humans , Meninges/pathology , Nerve Regeneration/physiology , Spinal Cord Injuries/pathology
10.
Cells ; 10(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34359956

ABSTRACT

Multiple sclerosis (MS) is a neuroimmune disorder characterized by inflammation, CNS demyelination, and progressive neurodegeneration. Chronic MS patients exhibit impaired remyelination capacity, partly due to the changes that oligodendrocyte precursor cells (OPCs) undergo in response to the MS lesion environment. The cytokine tumor necrosis factor (TNF) is present in the MS-affected CNS and has been implicated in disease pathophysiology. Of the two active forms of TNF, transmembrane (tmTNF) and soluble (solTNF), tmTNF signals via TNFR2 mediating protective and reparative effects, including remyelination, whereas solTNF signals predominantly via TNFR1 promoting neurotoxicity. To better understand the mechanisms underlying repair failure in MS, we investigated the cellular responses of OPCs to inflammatory exposure and the specific role of TNFR2 signaling in their modulation. Following treatment of cultured OPCs with IFNγ, IL1ß, and TNF, we observed, by RNA sequencing, marked inflammatory and immune activation of OPCs, accompanied by metabolic changes and dysregulation of their proliferation and differentiation programming. We also established the high likelihood of cell-cell interaction between OPCs and microglia in neuroinflammatory conditions, with OPCs able to produce chemokines that can recruit and activate microglia. Importantly, we showed that these functions are exacerbated when TNFR2 is ablated. Together, our data indicate that neuroinflammation leads OPCs to shift towards an immunomodulatory phenotype while diminishing their capacity to proliferate and differentiate, thus impairing their repair function. Furthermore, we demonstrated that TNFR2 plays a key role in this process, suggesting that boosting TNFR2 activation or its downstream signals could be an effective strategy to restore OPC reparative capacity in demyelinating disease.


Subject(s)
Cell Differentiation/physiology , Immunomodulation/immunology , Oligodendrocyte Precursor Cells/cytology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Remyelination/physiology , Animals , Cell Communication/immunology , Inflammation/immunology , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34132743

ABSTRACT

The wound healing process that occurs after spinal cord injury is critical for maintaining tissue homeostasis and limiting tissue damage, but eventually results in a scar-like environment that is not conducive to regeneration and repair. A better understanding of this dichotomy is critical to developing effective therapeutics that target the appropriate pathobiology, but a major challenge has been the large cellular heterogeneity that results in immensely complex cellular interactions. In this study, we used single-cell RNA sequencing to assess virtually all cell types that comprise the mouse spinal cord injury site. In addition to discovering novel subpopulations, we used expression values of receptor-ligand pairs to identify signaling pathways that are predicted to regulate specific cellular interactions during angiogenesis, gliosis, and fibrosis. Our dataset is a valuable resource that provides novel mechanistic insight into the pathobiology of not only spinal cord injury but also other traumatic disorders of the CNS.


Subject(s)
Cell Communication , Single-Cell Analysis , Spinal Cord Injuries/pathology , Angiopoietins/metabolism , Animals , Astrocytes/metabolism , Chemotaxis , Female , Fibroblasts/metabolism , Fibrosis , Gliosis/complications , Gliosis/pathology , Inflammation/pathology , Interleukin-6/metabolism , Ligands , Macrophages/pathology , Mice, Inbred C57BL , Myeloid Cells/pathology , Neuroglia/pathology , Oncostatin M/metabolism , Receptors, Oncostatin M/metabolism , Signal Transduction , Spinal Cord Injuries/complications , Spinal Cord Injuries/immunology , Time Factors , Transcriptome/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Mol Neurobiol ; 58(4): 1392-1403, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33184784

ABSTRACT

Traumatic optic neuropathy leads to bidirectional degeneration of retinal ganglion cells and axons and results in optic nerve scaring, which inhibits the regeneration of damaged axons. Compared with its glial counterpart, the fibrotic response causing nerve scar tissue is poorly permissive to axonal regeneration. Using collagen1α1-GFP reporter mice, we characterize the development of fibrotic scar formation following optic nerve crush injury. We observe that perivascular collagen1α1 cells constitute a major cellular component of the fibrotic scar. We demonstrate that extracellular molecules and monocytes are key factors contributing to the pathogenesis of optic nerve fibrotic scar formation, with a previously unrecognized encapsulation of this scar. We also characterize the distribution of collagen1α1 cells in the retina after optic nerve crush injury based on in vivo and whole-mount retinal imaging. Our results identify collagen1α1 cells as a major component of fibrotic scarring following ONC and are a potential molecular target for promoting axonal regeneration after optic nerve injury.


Subject(s)
Crush Injuries/pathology , Fibroblasts/pathology , Nerve Crush , Optic Nerve Injuries/pathology , Optic Nerve/pathology , Animals , Cell Count , Cicatrix/pathology , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Fibrosis , Macrophages/pathology , Mice, Transgenic , Microglia/pathology , Monocytes/pathology , Neuroglia/pathology , Pericytes/metabolism , Pericytes/pathology
14.
Front Cell Neurosci ; 14: 576037, 2020.
Article in English | MEDLINE | ID: mdl-33192321

ABSTRACT

The lack of effective treatments for most neurological diseases has prompted the search for novel therapeutic options. Interestingly, neuroinflammation is emerging as a common feature to target in most CNS pathologies. Recent studies suggest that targeted delivery of small molecules to reduce neuroinflammation can be beneficial. However, suboptimal drug delivery to the CNS is a major barrier to modulate inflammation because neurotherapeutic compounds are currently being delivered systemically without spatial or temporal control. Emerging nanomaterial technologies are providing promising and superior tools to effectively access neuropathological tissue in a controlled manner. Here we highlight recent advances in nanomaterial technologies for drug delivery to the CNS. We propose that state-of-the-art nanoparticle drug delivery platforms can significantly impact local CNS bioavailability of pharmacological compounds and treat neurological diseases.

15.
Adv Mater ; 32(43): e2002578, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32893402

ABSTRACT

Central nervous system (CNS) injuries are often debilitating, and most currently have no cure. This is due to the formation of a neuroinhibitory microenvironment at injury sites, which includes neuroinflammatory signaling and non-permissive extracellular matrix (ECM) components. To address this challenge, a viscous interfacial self-assembly approach, to generate a bioinspired hybrid 3D porous nanoscaffold platform for delivering anti-inflammatory molecules and establish a favorable 3D-ECM environment for the effective suppression of the neuroinhibitory microenvironment, is developed. By tailoring the structural and biochemical properties of the 3D porous nanoscaffold, enhanced axonal growth from the dual-targeting therapeutic strategy in a human induced pluripotent stem cell (hiPSC)-based in vitro model of neuroinflammation is demonstrated. Moreover, nanoscaffold-based approaches promote significant axonal growth and functional recovery in vivo in a spinal cord injury model through a unique mechanism of anti-inflammation-based fibrotic scar reduction. Given the critical role of neuroinflammation and ECM microenvironments in neuroinhibitory signaling, the developed nanobiomaterial-based therapeutic intervention may pave a new road for treating CNS injuries.


Subject(s)
Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cellular Microenvironment/drug effects , Central Nervous System/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacology , Nanostructures/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Axons/drug effects , Axons/metabolism , Biomimetic Materials/therapeutic use , Drug Carriers/therapeutic use , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Porosity , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
16.
Ocul Immunol Inflamm ; 28(5): 833-838, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-31418636

ABSTRACT

PURPOSE: We performed a meta-analysis to evaluate the effect of uveitis treatment on glaucoma drainage implant surgical outcomes. METHODS: We included 16 articles in the meta-analysis. Two groups were defined based on medical therapy of uveitis: Group 1: poorly controlled uveitis, and Group 2: well-controlled uveitis including use of immunomodulatory medications. RESULTS: The two groups were similar in comparisons of follow-up time, age, gender, and etiology of uveitis. Meta-analysis demonstrated significantly greater success in Group 2 (95.1%) compared to Group 1 (81.6%) at 1 year after glaucoma drainage implant surgery (P = .001). The final success was significantly greater (P 0.014) in group 2 compared with group 1 (86.1% and 74.3%, respectively). CONCLUSION: Surgical success was significantly higher in uveitic glaucoma patients treated with more intensive immunosuppressive therapy before and after glaucoma drainage implant surgery. The level of control of uveitis perioperatively appears to influence glaucoma drainage implant surgery outcomes.


Subject(s)
Glaucoma Drainage Implants , Glaucoma/physiopathology , Glaucoma/surgery , Immunomodulation , Intraocular Pressure/physiology , Uveitis/drug therapy , Adult , Female , Follow-Up Studies , Glaucoma/etiology , Humans , Male , Middle Aged , Treatment Outcome , Uveitis/complications , Visual Acuity/physiology
17.
Neurobiol Dis ; 134: 104674, 2020 02.
Article in English | MEDLINE | ID: mdl-31731043

ABSTRACT

Remyelination failure is a crucial component of disease progression in the autoimmune demyelinating disease Multiple Sclerosis (MS). The regenerative capacity of oligodendrocyte progenitor cells (OPCs) to replace myelinating oligodendrocytes is likely influenced by many aspects of the lesion environment including inflammatory signaling and extracellular matrix (ECM) deposition. These features of MS lesions are typically attributed to infiltrating leukocytes and reactive astrocytes. Here we demonstrate that fibroblasts also contribute to the inhibitory environment in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Using Col1α1GFP transgenic mice, we show that perivascular fibroblasts are activated in the spinal cord at EAE onset, and infiltrate the parenchyma by the peak of behavioral deficits where they are closely associated with areas of demyelination, myeloid cell accumulation, and ECM deposition. We further show that both fibroblast conditioned media and fibroblast ECM inhibit the differentiation of OPCs into mature oligodendrocytes. Taken together, our results indicate that the fibrotic scar is a major component of EAE pathology that leads to an inhibitory environment for remyelination, thus raising the possibility that anti-fibrotic mechanisms may serve as novel therapeutic targets for MS.


Subject(s)
Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/pathology , Oligodendroglia/pathology , Oligodendroglia/physiology , Spinal Cord/pathology , Animals , Fibroblasts/pathology , Fibrosis , Male , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , White Matter/pathology
18.
Nat Commun ; 10(1): 3028, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292434

ABSTRACT

Cerebellar neuronal progenitors undergo a series of divisions before irreversibly exiting the cell cycle and differentiating into neurons. Dysfunction of this process underlies many neurological diseases including ataxia and the most common pediatric brain tumor, medulloblastoma. To better define the pathways controlling the most abundant neuronal cells in the mammalian cerebellum, cerebellar granule cell progenitors (GCPs), we performed RNA-sequencing of GCPs exiting the cell cycle. Time-series modeling of GCP cell cycle exit identified downregulation of activity of the epigenetic reader protein Brd4. Brd4 binding to the Gli1 locus is controlled by Casein Kinase 1δ (CK1 δ)-dependent phosphorylation during GCP proliferation, and decreases during GCP cell cycle exit. Importantly, conditional deletion of Brd4 in vivo in the developing cerebellum induces cerebellar morphological deficits and ataxia. These studies define an essential role for Brd4 in cerebellar granule cell neurogenesis and are critical for designing clinical trials utilizing Brd4 inhibitors in neurological indications.


Subject(s)
Cerebellar Ataxia/genetics , Cerebellar Cortex/growth & development , Neural Stem Cells/physiology , Neurogenesis/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Casein Kinase Idelta , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cerebellar Ataxia/pathology , Cerebellar Cortex/cytology , Cerebellar Cortex/pathology , Disease Models, Animal , Down-Regulation , Humans , Mice , Mice, Knockout , Neurons/physiology , Nuclear Proteins/genetics , Phosphorylation/physiology , Primary Cell Culture , Transcription Factors/genetics , Zinc Finger Protein GLI1/metabolism
19.
Acta Neuropathol ; 137(5): 785-797, 2019 05.
Article in English | MEDLINE | ID: mdl-30929040

ABSTRACT

Virtually all phases of spinal cord injury pathogenesis, including inflammation, cell proliferation and differentiation, as well as tissue remodeling, are mediated in part by infiltrating monocyte-derived macrophages. It is now clear that these infiltrating macrophages have distinct functions from resident microglia and are capable of mediating both harmful and beneficial effects after injury. These divergent effects have been largely attributed to environmental cues, such as specific cytokines, that influence the macrophage polarization state. In this review, we also consider the possibility that different macrophage origins, including the spleen, bone marrow, and local self-renewal, may also affect macrophage fate, and ultimately their function that contribute to the complex pathobiology of spinal cord injury.


Subject(s)
Macrophages/pathology , Macrophages/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Humans
20.
Acta Neuropathol ; 137(5): 799-800, 2019 May.
Article in English | MEDLINE | ID: mdl-31011858

ABSTRACT

The original version of the article contains a labeling error in Fig. 2. The boxed molecular description of pro-inflammatory and anti-inflammatory macrophages were switched. Ly6CHi, Cx3Cr1Lo, Ccr2Hi should have been associated with pro-inflammatory macrophages on the left, and Ly6CLo, Cx3Cr1Hi, Ccr2Lo should have been associated with anti-inflammatory macrophages on the right.

SELECTION OF CITATIONS
SEARCH DETAIL
...