Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 20(1): 133, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37259118

ABSTRACT

Traumatic brain injury (TBI) remains a major cause of death and severe disability worldwide. We found previously that treatment with exogenous naïve B cells was associated with structural and functional neuroprotection after TBI. Here, we used a mouse model of unilateral controlled cortical contusion TBI to investigate cellular mechanisms of immunomodulation associated with intraparenchymal delivery of mature naïve B lymphocytes at the time of injury. Exogenous B cells showed a complex time-dependent response in the injury microenvironment, including significantly increased expression of IL-10, IL-35, and TGFß, but also IL-2, IL-6, and TNFα. After 10 days in situ, B cell subsets expressing IL-10 or TGFß dominated. Immune infiltration into the injury predominantly comprised myeloid cells, and B cell treatment did not alter overall numbers of infiltrating cells. In the presence of B cells, significantly more infiltrating myeloid cells produced IL-10, TGFß, and IL-35, and fewer produced TNFα, interferon-γ and IL-6 as compared to controls, up to 2 months post-TBI. B cell treatment significantly increased the proportion of CD206+ infiltrating monocytes/macrophages and reduced the relative proportion of activated microglia starting at 4 days and up to 2 months post-injury. Ablation of peripheral monocytes with clodronate liposomes showed that infiltrating peripheral monocytes/macrophages are required for inducing the regulatory phenotype in exogenous B cells. Reciprocally, B cells specifically reduced the expression of inflammatory cytokines in infiltrating Ly6C+ monocytes/macrophages. These data support the hypothesis that peripheral myeloid cells, particularly infiltrating monocyte/macrophages, are key mediators of the neuroprotective immunomodulatory effects observed after B cell treatment.


Subject(s)
Brain Injuries, Traumatic , Neuroprotective Agents , Mice , Animals , Interleukin-10/metabolism , Tumor Necrosis Factor-alpha/metabolism , Neuroprotection , Interleukin-6/metabolism , Brain Injuries, Traumatic/metabolism , Myeloid Cells/metabolism , Immunomodulation , Neuroprotective Agents/pharmacology , Transforming Growth Factor beta/metabolism , B-Lymphocytes/metabolism , Mice, Inbred C57BL , Microglia/metabolism
2.
Neurobiol Dis ; 180: 106077, 2023 05.
Article in English | MEDLINE | ID: mdl-36914074

ABSTRACT

Acute injury to the central nervous system (CNS) remains a complex and challenging clinical need. CNS injury initiates a dynamic neuroinflammatory response, mediated by both resident and infiltrating immune cells. Following the primary injury, dysregulated inflammatory cascades have been implicated in sustaining a pro-inflammatory microenvironment, driving secondary neurodegeneration and the development of lasting neurological dysfunction. Due to the multifaceted nature of CNS injury, clinically effective therapies for conditions such as traumatic brain injury (TBI), spinal cord injury (SCI), and stroke have proven challenging to develop. No therapeutics that adequately address the chronic inflammatory component of secondary CNS injury are currently available. Recently, B lymphocytes have gained increasing appreciation for their role in maintaining immune homeostasis and regulating inflammatory responses in the context of tissue injury. Here we review the neuroinflammatory response to CNS injury with particular focus on the underexplored role of B cells and summarize recent results on the use of purified B lymphocytes as a novel immunomodulatory therapeutic for tissue injury, particularly in the CNS.


Subject(s)
Brain Injuries, Traumatic , Spinal Cord Injuries , Humans , Central Nervous System , Spinal Cord Injuries/drug therapy , Inflammation , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/complications , B-Lymphocytes
3.
Front Cell Neurosci ; 17: 1329823, 2023.
Article in English | MEDLINE | ID: mdl-38269112

ABSTRACT

B lymphocytes are immune cells studied predominantly in the context of peripheral humoral immune responses against pathogens. Evidence has been accumulating in recent years on the diversity of immunomodulatory functions that B cells undertake, with particular relevance for pathologies of the central nervous system (CNS). This review summarizes current knowledge on B cell populations, localization, infiltration mechanisms, and function in the CNS and associated tissues. Acute and chronic neurodegenerative pathologies are examined in order to explore the complex, and sometimes conflicting, effects that B cells can have in each context, with implications for disease progression and treatment outcomes. Additional factors such as aging modulate the proportions and function of B cell subpopulations over time and are also discussed in the context of neuroinflammatory response and disease susceptibility. A better understanding of the multifactorial role of B cell populations in the CNS may ultimately lead to innovative therapeutic strategies for a variety of neurological conditions.

4.
Article in English | MEDLINE | ID: mdl-36445471

ABSTRACT

In adult mammals, spontaneous repair after spinal cord injury (SCI) is severely limited. By contrast, teleost fish successfully regenerate injured axons and produce new neurons from adult neural stem cells after SCI. The molecular mechanisms underlying this high regenerative capacity are largely unknown. The present study addresses this gap by examining the temporal dynamics of proteome changes in response to SCI in the brown ghost knifefish (Apteronotus leptorhynchus). Two-dimensional difference gel electrophoresis (2D DIGE) was combined with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and tandem mass spectrometry (MS/MS) to collect data during early (1 day), mid (10 days), and late (30 days) phases of regeneration following caudal amputation SCI. Forty-two unique proteins with significant differences in abundance between injured and intact control samples were identified. Correlation analysis uncovered six clusters of spots with similar expression patterns over time and strong conditional dependences, typically within functional families or between isoforms. Significantly regulated proteins were associated with axon development and regeneration; proliferation and morphogenesis; neuronal differentiation and re-establishment of neural connections; promotion of neuroprotection, redox homeostasis, and membrane repair; and metabolism or energy supply. Notably, at all three time points examined, significant regulation of proteins involved in inflammatory responses was absent.


Subject(s)
Gymnotiformes , Spinal Cord Injuries , Animals , Proteomics , Nerve Regeneration/physiology , Tandem Mass Spectrometry , Spinal Cord/metabolism , Gymnotiformes/physiology , Fishes , Mammals
6.
Nat Commun ; 13(1): 181, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013299

ABSTRACT

Diabetic foot ulceration (DFU) is a devastating complication of diabetes whose pathogenesis remains incompletely understood. Here, we profile 174,962 single cells from the foot, forearm, and peripheral blood mononuclear cells using single-cell RNA sequencing. Our analysis shows enrichment of a unique population of fibroblasts overexpressing MMP1, MMP3, MMP11, HIF1A, CHI3L1, and TNFAIP6 and increased M1 macrophage polarization in the DFU patients with healing wounds. Further, analysis of spatially separated samples from the same patient and spatial transcriptomics reveal preferential localization of these healing associated fibroblasts toward the wound bed as compared to the wound edge or unwounded skin. Spatial transcriptomics also validates our findings of higher abundance of M1 macrophages in healers and M2 macrophages in non-healers. Our analysis provides deep insights into the wound healing microenvironment, identifying cell types that could be critical in promoting DFU healing, and may inform novel therapeutic approaches for DFU treatment.


Subject(s)
Diabetes Mellitus/genetics , Diabetic Foot/genetics , Fibroblasts/metabolism , Macrophages/metabolism , Transcriptome , Wound Healing/genetics , Biomarkers/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Foot/metabolism , Diabetic Foot/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/pathology , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/pathology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 11/genetics , Matrix Metalloproteinase 11/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Single-Cell Analysis/methods , Skin/metabolism , Skin/pathology , Exome Sequencing
7.
FASEB J ; 35(12): e22019, 2021 12.
Article in English | MEDLINE | ID: mdl-34792819

ABSTRACT

Exogenously applied mature naïve B220+ /CD19+ /IgM+ /IgD+ B cells are strongly protective in the context of tissue injury. However, the mechanisms by which B cells detect tissue injury and aid repair remain elusive. Here, we show in distinct models of skin and brain injury that MyD88-dependent toll-like receptor (TLR) signaling through TLR2/6 and TLR4 is essential for the protective benefit of B cells in vivo, while B cell-specific deletion of MyD88 abrogated this effect. The B cell response to injury was multi-modal with simultaneous production of both regulatory cytokines, such as IL-10, IL-35, and transforming growth factor beta (TGFß), and inflammatory cytokines, such as tumor necrosis factor alpha (TNFα), IL-6, and interferon gamma. Cytometry analysis showed that this response was time and environment-dependent in vivo, with 20%-30% of applied B cells adopting an immune modulatory phenotype with high co-expression of anti- and pro-inflammatory cytokines after 18-48 h at the injury site. B cell treatment reduced the expression of TNFα and increased IL-10 and TGFß in infiltrating immune cells and fibroblasts at the injury site. Proteomic analysis further showed that B cells have a complex time-dependent homeostatic effect on the injured microenvironment, reducing the expression of inflammation-associated proteins, and increasing proteins associated with proliferation, tissue remodeling, and protection from oxidative stress. These findings chart and validate a first mechanistic understanding of the effects of B cells as an immunomodulatory cell therapy in the context of tissue injury.


Subject(s)
B-Lymphocytes/physiology , Brain Injuries/prevention & control , Cytokines/metabolism , Myeloid Differentiation Factor 88/physiology , Skin/immunology , Wound Healing , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Interleukin-10/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Skin/injuries , Skin/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Cell Mol Neurobiol ; 40(5): 695-710, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31808010

ABSTRACT

Cathepsin K deficiency in male mice (Ctsk-/-) results in decreased numbers of hippocampal astrocytes and altered neuronal patterning as well as learning and memory deficits. Additionally, cathepsin K carries essential roles in the thyroid gland where it contributes to the liberation of thyroid hormones (TH). Because TH are essential for brain development, in particular for the cerebellum, we investigated whether cathepsin K's function in the thyroid is directly linked to the brain phenotype of Ctsk-/- mice. Serum levels of thyroid stimulating hormone, brain concentrations of free TH, and deiodinase 2 (Dio2) activity in brain parenchyma as well as cerebellar development were comparable in Ctsk-/- and WT animals, suggesting regular thyroid states and TH metabolism. Despite unaltered transcript levels, protein expression of two TH transporters was enhanced in specific brain regions in Ctsk-/- mice, suggesting altered TH supply to these regions. Thyrotropin releasing hormone (Trh) mRNA levels were enhanced threefold in the hippocampus of Ctsk-/- mice. In the striatum of Ctsk-/- mice the mRNA for Dio2 and hairless were approximately 1.3-fold enhanced, while mRNA levels for monocarboxylate transporter 8 and Trh were reduced to 60% and 40%, respectively, pointing to altered striatal physiology. We conclude that the role of cathepsin K in the thyroid gland is not directly associated with its function in the central nervous system (CNS) of mice. Future studies will show whether the brain region-specific alterations in Trh mRNA may eventually result in altered neuroprotection that could explain the neurobehavioral defects of Ctsk-/- mice.


Subject(s)
Cathepsin K/physiology , Central Nervous System/enzymology , Thyroid Gland/enzymology , Animals , Cathepsin K/genetics , Cerebellum/enzymology , Cerebellum/growth & development , Male , Mice , Mice, Knockout , RNA, Messenger/analysis , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
10.
J Neurotrauma ; 36(17): 2579-2589, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30997843

ABSTRACT

Cerebral contusion causes neurological dysfunction mediated in part by inflammatory responses to injury. B lymphocytes are dynamic regulators of the immune system that have not been systematically studied in traumatic brain injury (TBI). We showed previously that topically applied mature B cells have immunomodulatory properties and strongly promote tissue regeneration, including cutaneous nerve growth, in acute and chronic skin wounds. Using a mouse controlled cortical impact (CCI) model, we assessed a possible beneficial role of exogenously applied B cells on histopathological and functional outcome after TBI. Mice were injected intraparenchymally at the lesion site with 2 × 106 mature naïve syngeneic splenic B cells, then subjected to CCI. Control CCI mice received equal numbers of T cells or saline, and sham-injured mice (craniotomy only) were given B cells or saline. Sham-injured groups performed similarly in motor and learning tests. Injured mice administered B cells showed significantly improved post-injury rotarod, Y maze, and Morris water maze (MWM) performance compared with saline- or T-cell-treated CCI groups. Moreover, lesion volume in mice treated with B cells was significantly reduced by 40% at 35 days post-TBI compared with saline and T cell controls, and astrogliosis and microglial activation were decreased. In vivo tracking of exogenous B cells showed that they have a limited life span of approximately 14 days in situ and do not appear to proliferate. The data suggest proof of principle that local administration of B lymphocytes may represent a therapeutic option for treatment of cerebral contusion, especially when clinical management involves procedures that allow access to the injury site.


Subject(s)
B-Lymphocytes/transplantation , Brain Contusion/pathology , Brain Contusion/physiopathology , Recovery of Function/physiology , Animals , Male , Mice , Mice, Inbred C57BL
11.
Am J Transplant ; 19(7): 1930-1940, 2019 07.
Article in English | MEDLINE | ID: mdl-30748094

ABSTRACT

Pancreatic ß-cell replacement by islet transplantation for the treatment of type 1 diabetes (T1D) is currently limited by donor tissue scarcity and the requirement for lifelong immunosuppression. The advent of in vitro differentiation protocols for generating functional ß-like cells from human pluripotent stem cells, also referred to as SC-ß cells, could eliminate these obstacles. To avoid the need for immunosuppression, alginate-microencapsulation is widely investigated as a safe path to ß-cell replacement. Nonetheless, inflammatory foreign body responses leading to pericapsular fibrotic overgrowth often causes microencapsulated islet-cell death and graft failure. Here we used a novel approach to evade the pericapsular fibrotic response to alginate-microencapsulated SC-ß cells; an immunomodulatory chemokine, CXCL12, was incorporated into clinical grade sodium alginate to microencapsulate SC-ß cells. CXCL12 enhanced glucose-stimulated insulin secretion activity of SC-ß cells and induced expression of genes associated with ß-cell function in vitro. SC-ß cells co-encapsulated with CXCL12 showed enhanced insulin secretion in diabetic mice and accelerated the normalization of hyperglycemia. Additionally, SC-ß cells co-encapsulated with CXCL12 evaded the pericapsular fibrotic response, resulting in long-term functional competence and glycemic correction (>150 days) without systemic immunosuppression in immunocompetent C57BL/6 mice. These findings lay the groundwork for further preclinical translation of this approach into large animal models of T1D.


Subject(s)
Alginates/chemistry , Chemokine CXCL12/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Graft Survival , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation/methods , Stem Cells/cytology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Stem Cells/metabolism
12.
J Immunol ; 201(12): 3587-3603, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30420435

ABSTRACT

The treatment of skin with a low-power continuous-wave (CW) near-infrared (NIR) laser prior to vaccination is an emerging strategy to augment the immune response to intradermal vaccine, potentially substituting for chemical adjuvant, which has been linked to adverse effects of vaccines. This approach proved to be low cost, simple, small, and readily translatable compared with the previously explored pulsed-wave medical lasers. However, little is known on the mode of laser-tissue interaction eliciting the adjuvant effect. In this study, we sought to identify the pathways leading to the immunological events by examining the alteration of responses resulting from genetic ablation of innate subsets including mast cells and specific dendritic cell populations in an established model of intradermal vaccination and analyzing functional changes of skin microcirculation upon the CW NIR laser treatment in mice. We found that a CW NIR laser transiently stimulates mast cells via generation of reactive oxygen species, establishes an immunostimulatory milieu in the exposed tissue, and provides migration cues for dermal CD103+ dendritic cells without inducing prolonged inflammation, ultimately augmenting the adaptive immune response. These results indicate that use of an NIR laser with distinct wavelength and power is a safe and effective tool to reproducibly modulate innate programs in skin. These mechanistic findings would accelerate the clinical translation of this technology and warrant further explorations into the broader application of NIR lasers to the treatment of immune-related skin diseases.


Subject(s)
Dendritic Cells/immunology , Laser Therapy/methods , Mast Cells/immunology , Skin/immunology , Vaccines/immunology , Adaptive Immunity , Adjuvants, Immunologic , Animals , Cell Movement , Cells, Cultured , Female , Immunity, Innate , Immunization , Infrared Rays , Mice , Mice, Inbred C57BL , Radiation Exposure , Reactive Oxygen Species/metabolism , Skin/radiation effects
13.
Wound Repair Regen ; 25(5): 774-791, 2017 09.
Article in English | MEDLINE | ID: mdl-28922523

ABSTRACT

Chronic wounds affect 12-15% of patients with diabetes and are associated with a drastic decrease in their quality of life. Here, we demonstrate that purified mature naive B220+ /CD19+ /IgM+ /IgD+ B cells improve healing of acute and diabetic murine wounds after a single topical application. B cell treatment significantly accelerated acute wound closure by 2-3 days in wild-type mice and 5-6 days in obese diabetic mice. The treatment led to full closure in 43% of chronic diabetic wounds, as compared to only 5% in saline-treated controls. Applying equivalent numbers of T cells or disrupted B cells failed to reproduce these effects, indicating that live B cells mediated pro-healing responses. Topically applied B cell treatment was associated with significantly reduced scar size, increased collagen deposition and maturation, enhanced angiogenesis, and increased nerve growth into and under the healing wound. ß-III tubulin+ nerve endings in scars of wounds treated acutely with B cells showed increased relative expression of growth-associated protein 43. The improved healing associated with B cell treatment was supported by significantly increased fibroblast proliferation and decreased apoptosis in the wound bed and edges, altered kinetics of neutrophil infiltration, as well as an increase in TGF-ß and a significant reduction in MMP2 expression in wound granulation tissue. Our findings indicate that the timeline and efficacy of wound healing can be experimentally manipulated through the direct application of mature, naive B cells, which effectively modify the balance of mature immune cell populations within the wound microenvironment and accelerate the healing process.


Subject(s)
B-Lymphocytes , Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus, Experimental/complications , Skin Diseases/therapy , Skin/pathology , Wound Healing/immunology , Acute Disease , Animals , Biopsy , Cell Survival , Chronic Disease , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Skin/immunology , Skin Diseases/etiology , Skin Diseases/pathology
14.
Dev Neurobiol ; 77(11): 1269-1307, 2017 11.
Article in English | MEDLINE | ID: mdl-28707354

ABSTRACT

The knifefish Apteronotus leptorhynchus exhibits indeterminate growth throughout adulthood. This phenomenon extends to the spinal cord, presumably through the continuous addition of new neurons and glial cells. However, little is known about the developmental dynamics of cells added during adult growth. The present work characterizes the structural and functional development of the adult spinal cord in this model organism through a comprehensive quantitative analysis of the spatial and temporal dynamics of new cells at various developmental stages. This analysis, based on a novel statistical mapping approach, revealed within the adult spinal cord a wide distribution of both mitotically active and quiescent Sox2-expressing stem/progenitor cells (SPCs). While such cells are particularly concentrated within the ependymal layer near the central canal, the majority of them reside in the parenchyma, resembling the distribution of SPCs observed in the mammalian spinal cord. The active SPCs in the adult knifefish spinal cord give rise to transit amplifying progenitor cells that undergo a few additional mitotic divisions before developing into Hu C/D+ neurons and S100+ glial cells. There is no evidence of long-distance migration of the newborn cells. The persistence of cell proliferation and differentiation, combined with low levels of apoptosis, leads to a continuous addition of cells to the existing tissue. Newly generated neurons have functional and behavioral relevance, as indicated by the integration of axons of new electromotor neurons into the electric organ of these weakly electric fish. This results in a gradual increase in the amplitude of the electric organ discharge during adult development. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1269-1307, 2017.


Subject(s)
Cell Differentiation/physiology , Multipotent Stem Cells/physiology , Neurogenesis/physiology , Spinal Cord/cytology , Spinal Cord/growth & development , Animals , Bromodeoxyuridine/metabolism , Cell Count , ELAV Proteins/metabolism , Electric Fish , Electric Organ/cytology , Electric Organ/physiology , Female , Fluorescein/metabolism , Glutamate-Ammonia Ligase/metabolism , Histones/metabolism , Male , Models, Anatomic , Nerve Tissue Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , SOXB1 Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
15.
J Immunol ; 199(4): 1319-1332, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28710250

ABSTRACT

Brief exposure of skin to near-infrared (NIR) laser light has been shown to augment the immune response to intradermal vaccination and thus act as an immunologic adjuvant. Although evidence indicates that the NIR laser adjuvant has the capacity to activate innate subsets including dendritic cells (DCs) in skin as conventional adjuvants do, the precise immunological mechanism by which the NIR laser adjuvant acts is largely unknown. In this study we sought to identify the cellular target of the NIR laser adjuvant by using an established mouse model of intradermal influenza vaccination and examining the alteration of responses resulting from genetic ablation of specific DC populations. We found that a continuous wave (CW) NIR laser adjuvant broadly modulates migratory DC (migDC) populations, specifically increasing and activating the Lang+ and CD11b-Lang- subsets in skin, and that the Ab responses augmented by the CW NIR laser are dependent on DC subsets expressing CCR2 and Langerin. In comparison, a pulsed wave NIR laser adjuvant showed limited effects on the migDC subsets. Our vaccination study demonstrated that the efficacy of the CW NIR laser is significantly better than that of the pulsed wave laser, indicating that the CW NIR laser offers a desirable immunostimulatory microenvironment for migDCs. These results demonstrate the unique ability of the NIR laser adjuvant to selectively target specific migDC populations in skin depending on its parameters, and highlight the importance of optimization of laser parameters for desirable immune protection induced by an NIR laser-adjuvanted vaccine.


Subject(s)
Dendritic Cells/immunology , Influenza Vaccines/immunology , Infrared Rays , Lasers , Skin/immunology , Skin/radiation effects , Vaccination/methods , Adjuvants, Immunologic , Animals , Antigens, Surface/metabolism , Cell Movement , Dendritic Cells/physiology , Influenza Vaccines/administration & dosage , Injections, Intradermal , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Mice , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
16.
Article in English | MEDLINE | ID: mdl-27225982

ABSTRACT

Among the cellular processes that follow injury to the central nervous system, glial scar formation is thought to be one of the major factors that prevent regeneration. In regeneration-competent organisms, glial scar formation has been a matter of controversy. We addressed this issue by examining the glial population after spinal cord injury in a model of regeneration competency, the knifefish Apteronotus leptorhynchus. Analysis of spinal cord sections immunostained against the glial markers glial fibrillary acidic protein, vimentin, or chondroitin sulfate proteoglycan failed to produce any evidence for the formation of a glial scar in the area of the lesion at post-injury survival times ranging from 5 to 185 days. This result was independent of the lesion paradigm applied-amputation of the caudal part of the spinal cord or hemisection lesioning-and similar after examination of transverse and longitudinal sections. We hypothesize that the well-developed network of radial glia in both the intact and the injured spinal cord provides a support system for regeneration of tissue lost to injury. This glial network is likely also involved in the generation of new cells, as indicated by the large subset of glial fibrillary acidic protein-labeled glia that express the stem cell marker Sox2.


Subject(s)
Gymnotiformes/physiology , Neuroglia/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Regeneration/physiology , Animals , Disease Models, Animal , Fish Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis , Immunohistochemistry , Microscopy, Confocal , Microscopy, Fluorescence , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Neuroglia/pathology , SOXB1 Transcription Factors/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology
17.
BMC Genomics ; 16: 166, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25879418

ABSTRACT

BACKGROUND: The brown ghost knifefish (Apteronotus leptorhynchus) is a weakly electric teleost fish of particular interest as a versatile model system for a variety of research areas in neuroscience and biology. The comprehensive information available on the neurophysiology and neuroanatomy of this organism has enabled significant advances in such areas as the study of the neural basis of behavior, the development of adult-born neurons in the central nervous system and their involvement in the regeneration of nervous tissue, as well as brain aging and senescence. Despite substantial scientific interest in this species, no genomic resources are currently available. RESULTS: Here, we report the de novo assembly and annotation of the A. leptorhynchus transcriptome. After evaluating several trimming and transcript reconstruction strategies, de novo assembly using Trinity uncovered 42,459 unique contigs containing at least a partial protein-coding sequence based on alignment to a reference set of known Actinopterygii sequences. As many as 11,847 of these contigs contained full or near-full length protein sequences, providing broad coverage of the proteome. A variety of non-coding RNA sequences were also identified and annotated, including conserved long intergenic non-coding RNA and other long non-coding RNA observed previously to be expressed in adult zebrafish (Danio rerio) brain, as well as a variety of miRNA, snRNA, and snoRNA. Shotgun proteomics confirmed translation of open reading frames from over 2,000 transcripts, including alternative splice variants. Assignment of tandem mass spectra was greatly improved by use of the assembly compared to databases of sequences from closely related organisms. The assembly and raw reads have been deposited at DDBJ/EMBL/GenBank under the accession number GBKR00000000. Tandem mass spectrometry data is available via ProteomeXchange with identifier PXD001285. CONCLUSIONS: Presented here is the first release of an annotated de novo transcriptome assembly from Apteronotus leptorhynchus, providing a broad overview of RNA expressed in central nervous system tissue. The assembly, which includes substantial coverage of a wide variety of both protein coding and non-coding transcripts, will allow the development of better tools to understand the mechanisms underlying unique characteristics of the knifefish model system, such as their tremendous regenerative capacity and negligible brain senescence.


Subject(s)
Central Nervous System/metabolism , Fishes/genetics , Proteomics , Transcriptome , Amino Acid Sequence , Animals , Brain/metabolism , Chromatography, High Pressure Liquid , Contig Mapping , Fishes/classification , Fishes/metabolism , Genome , Molecular Sequence Data , Proteome/analysis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sequence Alignment , Sequence Analysis, RNA , Spinal Cord/metabolism , Tandem Mass Spectrometry
18.
Mol Cell Neurosci ; 68: 9-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25827096

ABSTRACT

Matrix metalloproteinases (MMPs) are a family of highly conserved zinc-dependent proteases involved in both development and pathogenesis. The present study examines the role of MMP-2 (gelatinase A) and MMP-9 (gelatinase B) in adult neurogenesis, using the corpus cerebelli, a subdivision of the cerebellum, of knifefish (Apteronotus leptorhynchus) as a model system. Transcripts of five isoforms of these gelatinases were identified in the central nervous system of this species. Sequence similarity analysis and homology modeling indicated that functionally and structurally critical elements were highly conserved in knifefish gelatinases. Immunohistochemical staining revealed a differential distribution of MMP-2 and MMP-9 at both the cellular and subcellular level. MMP-2 expression was found mainly in Sox2-immunopositive stem/progenitor cells, both quiescent and mitotically active; and was localized in both the cytoplasmic compartment and the nucleus. By contrast, MMP-9 immunoreactivity was absent in neurogenic niches and displayed a more homogenous distribution, with low to moderate intensity levels, in the molecular and granular layers. MMP-9 expression appeared to be restricted to the extracellular space. In situ zymography indicated that gelatinase activity matched the cellular and subcellular distributions of the two MMPs. The observed patterns of gelatinase activity and expression support the hypothesis that MMP-2 is primarily involved in regulation of the activity of stem/progenitor cells that give rise to new granule neurons, whereas MMP-9 facilitates migration of the progeny of these cells by proteolysis of extracellular matrix proteins.


Subject(s)
Cerebellum/enzymology , Matrix Metalloproteinase 9/metabolism , Neurogenesis/physiology , Adult Stem Cells/physiology , Animals , Cell Movement/physiology , Cerebellum/cytology , Fishes , Gelatinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Matrix Metalloproteinase 2/metabolism , Models, Molecular , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Subcellular Fractions/enzymology , Vimentin/metabolism
19.
Exp Neurol ; 263: 272-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448008

ABSTRACT

Many disorders of the CNS are characterized by a massive loss of neurons. A promising therapeutic strategy to cure such conditions is based on the activation of endogenous stem cells. Implementation of this strategy will benefit from a better understanding of stem cell dynamics and the local CNS microenvironment in regeneration-competent vertebrate model systems. Using a spinal cord injury paradigm in zebrafish larvae, Briona and Dorsky (2014) have provided evidence for the existence of two distinct neural stem cell populations. One population has the characteristics of radial glia and expresses the homeobox transcription factor Dbx. The other lacks Dbx but expresses Olig2. These results are placed in the context of other studies that also support the notion of heterogeneity of adult stem cells in the CNS. The implication that differences among stem cell populations, in combination with specific factors from the local cellular microenvironment, might have a decisive impact on the fate choices of the new cells, is discussed. Reviewed evidence suggests that rather few modifications in the signaling pathways involved in the control of stem cell behavior have led, in the course of evolution, to the pronounced differences between mammals and regeneration-competent organisms. As a consequence, rather minor pharmacological manipulations may be sufficient to reactivate the hidden neurogenic potential of the mammalian CNS, and thus make it available for therapeutic applications.


Subject(s)
Nerve Regeneration/physiology , Neural Stem Cells/physiology , Neuroglia/physiology , Neurons/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord/physiopathology , Animals
20.
Dev Neurobiol ; 75(1): 39-65, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25044932

ABSTRACT

Adult neurogenesis has been described in dozens of brain regions in teleost fish, with the largest number of new neurons being generated in the cerebellum. Here, we characterized the cerebellar neural stem/progenitor cells (NSPCs) in the brown ghost knifefish (Apteronotus leptorhynchus), an established model system of adult neurogenesis. The majority of the new cerebellar cells arise from neurogenic niches located medially, at the interface of the dorsal/ventral molecular layers and the granular layer. NSPCs within these niches give rise to transit-amplifying progenitors which populate the molecular layer, where they continue to proliferate during their migration toward target areas in the granular layer. At any given time, the majority of proliferating cells are located in the molecular layer. Immunohistochemical staining revealed that the stem cell markers Sox2, Meis1/2/3, Islet1, and, to a lesser extent, Pax6, are widely expressed in all regions of the adult cerebellum. A large subpopulation of these NSPCs coexpress S100, GFAP, and/or vimentin, indicating astrocytic identity. This is further supported by the specific effect of the gliotoxin l-methionine sulfoximine, which leads to a targeted decrease in the number of GFAP+ cells that coexpress Sox2 or the proliferation marker PCNA. Pulse-chase analysis of the label size associated with new cells after administration of 5-bromo-2'-deoxyuridine demonstrated that, on average, two additional cell divisions occur after completion of the initial mitotic cycle. Overall numbers of NSPCs in the cerebellum niches increase consistently over time, presumably in parallel with the continuous growth of the brain.


Subject(s)
Adult Stem Cells/physiology , Cerebellum/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Adult Stem Cells/cytology , Animals , Cerebellum/cytology , Fishes , Neural Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...