Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1399989, 2024.
Article in English | MEDLINE | ID: mdl-38799448

ABSTRACT

Introduction: Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods: Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results: Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119 + P2ry12 + microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion: Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease.


Subject(s)
Brain , Microglia , Retina , Animals , Microglia/immunology , Microglia/metabolism , Mice , Retina/immunology , Retina/pathology , Brain/immunology , Brain/pathology , Brain/metabolism , Mice, Inbred C57BL , Lymphocytic choriomeningitis virus/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Single-Cell Analysis , CD8-Positive T-Lymphocytes/immunology , Transcriptome
2.
J Neuroinflammation ; 20(1): 238, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858232

ABSTRACT

BACKGROUND: Neovascular age-related macular degeneration causes vision loss from destructive angiogenesis, termed choroidal neovascularization (CNV). Cx3cr1-/- mice display alterations in non-classical monocytes and microglia with increased CNV size, suggesting that non-classical monocytes may inhibit CNV formation. NR4A1 is a transcription factor that is necessary for maturation of non-classical monocytes from classical monocytes. While Nr4a1-/- mice are deficient in non-classical monocytes, results are confounded by macrophage hyper-activation. Nr4a1se2/se2 mice lack a transcriptional activator, resulting in non-classical monocyte loss without macrophage hyper-activation. MAIN BODY: We subjected Nr4a1-/- and Nr4a1se2/se2 mice to the laser-induced CNV model and performed multi-parameter flow cytometry. We found that both models lack non-classical monocytes, but only Nr4a1-/- mice displayed increased CNV area. Additionally, CD11c+ macrophages were increased in Nr4a1-/- mice. Single-cell transcriptomic analysis uncovered that CD11c+ macrophages were enriched from Nr4a1-/- mice and expressed a pro-angiogenic transcriptomic profile that was disparate from prior reports of macrophage hyper-activation. CONCLUSIONS: These results suggest that non-classical monocytes are dispensable during CNV, and NR4A1 deficiency results in increased recruitment of pro-angiogenic macrophages.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Animals , Mice , Choroidal Neovascularization/genetics , Disease Models, Animal , Macrophages/physiology , Macular Degeneration/genetics , Mice, Inbred C57BL , Microglia , Monocytes
3.
Sci Adv ; 9(41): eadi8534, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37831763

ABSTRACT

Neovascular age-related macular degeneration (nAMD) is the leading cause of blindness in the developed world. Current therapy includes monthly intraocular injections of anti-VEGF antibodies, which are ineffective in up to one third of patients. Thrombospondin-1 (TSP1) inhibits angiogenesis via CD36 binding, and its down-regulated expression is negatively associated with the onset of nAMD. Here, we describe TSP1 mimetic protein-like polymers (TSP1 PLPs). TSP1 PLPs bind CD36 with high affinity, resist degradation, show prolonged intraocular half-lives (13.1 hours), have no toxicity at relevant concentrations in vivo (40 µM), and are more efficacious in ex vivo choroidal sprouting assays compared to the peptide sequence and Eylea (aflibercept), the current standard of care anti-VEGF treatment. Furthermore, PLPs exhibit superior in vivo efficacy in a mouse model for nAMD compared to control PLPs consisting of scrambled peptide sequences, using fluorescein angiography and immunofluorescence. Since TSP-1 inhibits angiogenesis by VEGF-dependent and independent mechanisms, TSP1 PLPs are a potential therapeutic for patients with anti-VEGF treatment-resistant nAMD.


Subject(s)
Macular Degeneration , Ranibizumab , Animals , Mice , Humans , Ranibizumab/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Thrombospondin 1/therapeutic use , Macular Degeneration/drug therapy , Peptides
4.
JCI Insight ; 8(7)2023 04 10.
Article in English | MEDLINE | ID: mdl-36821388

ABSTRACT

Patients with neovascular AMD (nAMD) suffer vision loss from destructive angiogenesis, termed choroidal neovascularization (CNV). Macrophages are found in CNV lesions from patients with nAMD. Additionally, Ccr2-/- mice, which lack classical monocyte-derived macrophages, show reduced CNV size. However, macrophages are highly diverse cells that can perform multiple functions. We performed single-cell RNA-Seq on immune cells from WT and Ccr2-/- eyes to uncover macrophage heterogeneity during the laser-induced CNV mouse model of nAMD. We identified 12 macrophage clusters, including Spp1+ macrophages. Spp1+ macrophages were enriched from WT lasered eyes and expressed a proangiogenic transcriptome via multiple pathways, including vascular endothelial growth factor signaling, endothelial cell sprouting, cytokine signaling, and fibrosis. Additionally, Spp1+ macrophages expressed the marker CD11c, and CD11c+ macrophages were increased by laser and present in CNV lesions. Finally, CD11c+ macrophage depletion reduced CNV size by 40%. These findings broaden our understanding of ocular macrophage heterogeneity and implicate CD11c+ macrophages as potential therapeutic targets for treatment-resistant patients with nAMD.


Subject(s)
Choroidal Neovascularization , Wet Macular Degeneration , Animals , Mice , Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Macrophages/metabolism , Vascular Endothelial Growth Factor A/metabolism , Visual Acuity , Wet Macular Degeneration/pathology , CD11c Antigen/metabolism
5.
J Neuroinflammation ; 19(1): 203, 2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35941655

ABSTRACT

BACKGROUND: Diabetic retinopathy and retinal vein occlusion are vision threatening retinal vascular diseases. Current first-line therapy targets the vascular component, but many patients are treatment-resistant due to unchecked inflammation. Non-invasive inflammatory imaging biomarkers are a significant unmet clinical need for patients. Imaging of macrophage-like cells on the surface of the retina using clinical optical coherence tomography (OCT) is an emerging field. These cells are increased in patients with retinal vascular disease, and could be a potential inflammatory biomarker. However, since OCT is limited by an axial resolution of 5-10 microns, the exact location and identity of these retinal cells is currently unknown. METHODS: We performed OCT followed by confocal immunofluorescence in wild-type mice to identify macrophages within 5-10 microns of the vitreoretinal interface. Next, we used Cx3cr1CreER/+; Rosa26zsGreen/+ mice to fate map retinal surface macrophages. Using confocal immunofluorescence of retinal sections and flatmounts, we quantified IBA1+Tmem119+CD169neg microglia, IBA1+Tmem119negCD169neg perivascular macrophages, and IBA1+Tmem119negCD169+ vitreal hyalocytes. Finally, we modeled neuroinflammation with CCL2 treatment and characterized retinal surface macrophages using flow cytometry, OCT, and confocal immunofluorescence. RESULTS: We were able to detect IBA1+ macrophages within 5-10 microns of the vitreoretinal interface in wild-type mice using OCT followed by confirmatory confocal immunofluorescence. Retinal surface macrophages were 83.5% GFP+ at Week 1 and 82.4% GFP+ at Week 4 using fate mapping mice. At steady state, these macrophages included 82% IBA1+Tmem119+CD169neg microglia, 9% IBA1+Tmem119negCD169+ vitreal hyalocytes, and 9% IBA1+Tmem119negCD169neg perivascular macrophages. After CCL2-driven neuroinflammation, many Ly6C+ cells were detectable on the retinal surface using OCT followed by confocal immunofluorescence. CONCLUSIONS: Macrophages within close proximity to the vitreoretinal interface are self-renewing cells, and predominantly microglia with minor populations of perivascular macrophages and vitreal hyalocytes at steady state. In the context of neuroinflammation, monocytes and monocyte-derived macrophages are a significant component of retinal surface macrophages. Human OCT-based imaging of retinal surface macrophages is a potential biomarker for inflammation during retinal vascular disease.


Subject(s)
Retinal Diseases , Retinal Vein Occlusion , Animals , Biomarkers , Disease Models, Animal , Humans , Inflammation/diagnostic imaging , Macrophages , Mice , Microglia
6.
Sci Rep ; 11(1): 18084, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508129

ABSTRACT

Neovascular age-related macular degeneration (nAMD) commonly causes vision loss from aberrant angiogenesis, termed choroidal neovascularization (CNV). Interleukin-6 (IL6) is a pro-inflammatory and pro-angiogenic cytokine that is correlated with AMD progression and nAMD activity. We hypothesize that anti-IL6 therapy is a potential nAMD therapeutic. We found that IL6 levels were increased after laser injury and expressed by macrophages. Il6-deficiency decreased laser-induced CNV area and exogenous IL6 addition increased choroidal sprouting angiogenesis. Il6-null mice demonstrated equally increased macrophage numbers as wildtype mice. At steady state, IL6R expression was detected on peripheral blood and ocular monocytes. After laser injury, the number of IL6R+Ly6C+ monocytes in blood and IL6R+ macrophages in the eye were increased. In human choroid, macrophages expressed IL6, IL6R, and IL6ST. Furthermore, IL6R+ macrophages displayed a transcriptional profile consistent with STAT3 (signal transducer and activator of transcription 3) activation and angiogenesis. Our data show that IL6 is both necessary and sufficient for choroidal angiogenesis. Macrophage-derived IL6 may stimulate choroidal angiogenesis via classical activation of IL6R+ macrophages, which then stimulate angiogenesis. Targeting IL6 or the IL6R could be an effective adjunctive therapy for treatment-resistant nAMD patients.


Subject(s)
Choroid/blood supply , Choroid/metabolism , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Animals , Biomarkers , Choroid/pathology , Choroidal Neovascularization/pathology , Choroidal Neovascularization/therapy , Disease Models, Animal , Disease Susceptibility , Humans , Mice , Monocytes/metabolism , Receptors, Interleukin-6/metabolism
7.
Sci Rep ; 11(1): 17254, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446787

ABSTRACT

Age-related macular degeneration (AMD) is genetically associated with complement. Dendritic cells (DCs) play key roles during innate and adaptive immunity, and express complement components and their receptors. We investigated ocular DC heterogeneity and the role of DCs in the laser-induced choroidal neovascularization (CNV) model. In order to determine the function of DCs, we used two models of DC deficiency: the Flt3-/- and Flt3l-/- mouse. We identified three types of ocular DCs: plasmacytoid DC, classical DC-1, and classical DC-2. At steady-state, classical DCs were found in the iris and choroid but were not detectable in the retina. Plasmacytoid DCs existed at very low levels in iris, choroid, and retina. After laser injury, the number of each DC subset was up-regulated in the choroid and retina. In Flt3-/- mice, we found reduced numbers of classical DCs at steady-state, but each DC subset equally increased after laser injury between wildtype and Flt3-/- mice. In Flt3l-/- mice, each DC subsets was severely reduced after laser injury. Neither Flt3-/- or Flt3l-/- mice demonstrated reduced CNV area compared to wildtype mice. DCs do not play any significant role during the laser-induced CNV model of neovascular AMD.


Subject(s)
Choroid/immunology , Choroidal Neovascularization/immunology , Dendritic Cells/immunology , Membrane Proteins/immunology , fms-Like Tyrosine Kinase 3/immunology , Animals , Choroid/blood supply , Choroidal Neovascularization/etiology , Choroidal Neovascularization/genetics , Female , Flow Cytometry/methods , Iris/blood supply , Iris/immunology , Lasers/adverse effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Retina/immunology , Visual Acuity/immunology , Wet Macular Degeneration/immunology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
8.
J Neuroinflammation ; 17(1): 341, 2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33187533

ABSTRACT

BACKGROUND: Neovascular age-related macular degeneration (nAMD) commonly causes vision loss from aberrant angiogenesis, termed choroidal neovascularization (CNV). Macrophages are heterogeneous cells that are necessary for experimental CNV, present in human CNV samples, and can display diverse functions, which are dependent upon both their origin and tissue microenvironment. Despite these associations, choroidal macrophage heterogeneity remains unexplored. METHODS: We performed multi-parameter flow cytometry on wildtype (WT) and Ccr2-/- mice after laser injury to identify macrophage subtypes, and determine which subsets originate from classical monocytes. To fate map tissue resident macrophages at steady state and after laser injury, we used the Cx3cr1CreER/+ ; Rosa26zsGFP/+ mouse model. We reanalyzed previously published single-cell RNA-seq of human choroid samples from healthy and nAMD patients to investigate human macrophage heterogeneity, disease association, and function. RESULTS: We identified 4 macrophage subsets in mice: microglia, MHCII+CD11c-, MHCII+CD11c+, and MHCII-. Microglia are tissue resident macrophages at steady state and unaffected by laser injury. At steady state, MHCII- macrophages are long lived, tissue resident macrophages, while MHCII+CD11c- and MHCII+CD11c+ macrophages are partially replenished from blood monocytes. After laser injury, MHCII+CD11c- macrophages are entirely derived from classical monocytes, MHCII- macrophages originate from classical monocytes (90%) and an expansion of tissue resident macrophages (10%), and MHCII+CD11c+ macrophages are derived from classical monocytes (70%), non-classical monocytes (10%), and an expansion of tissue resident macrophages (20%). Single-cell RNA-seq analysis of human choroid found 5 macrophage subsets: two MHCII+CD11C- and three MHCII+CD11C+ populations. One MHCII+CD11C+ subset was 78% derived from a patient with nAMD. Differential expression analysis identified up-regulation of pro-angiogenic gene expression in one MHCII+CD11C- and two MHCII+CD11C+ subsets, including the disease-associated cluster. The upregulated MHCII+CD11C- pro-angiogenic genes were unique compared to the increased MHCII+CD11C+ angiogenesis genes. CONCLUSIONS: Macrophage origin impacts heterogeneity at steady state and after laser injury in mice. Both mice and human patients demonstrate similar macrophage subtypes. Two discrete pro-angiogenic macrophage populations exist in the human choroid. Targeting specific, pro-angiogenic macrophage subsets is a potential novel therapeutic for nAMD.


Subject(s)
Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Genetic Heterogeneity , Macrophages/metabolism , Animals , Choroidal Neovascularization/pathology , Female , Laser Therapy/adverse effects , Macrophages/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
9.
J Vis Exp ; (160)2020 06 17.
Article in English | MEDLINE | ID: mdl-32628177

ABSTRACT

The innate immune system plays important roles in ocular pathophysiology including uveitis, diabetic retinopathy, and age-related macular degeneration. Innate immune cells, specifically mononuclear phagocytes, express overlapping cell surface markers, which makes identifying these populations a challenge. Multi-parameter flow cytometry allows for the simultaneous, quantitative analysis of multiple cell surface markers in order to differentiate monocytes, macrophages, microglia, and dendritic cells in mouse eyes. This protocol describes the enucleation of whole mouse eyes, ocular dissection, digestion into a single cell suspension, and staining of the single cell suspension for myeloid cell markers. Additionally, we explain the proper methods for determining voltages using single color controls and for delineating positive gates using fluorescence minus one controls. The major limitation of multi-parameter flow cytometry is the absence of tissue architecture. This limitation can be overcome by multi-parameter flow cytometry of individual ocular compartments or complimentary immunofluorescence staining. However, immunofluorescence is limited by its lack of quantitative analysis and reduced number of fluorophores on most microscopes. We describe the use of multi-parametric flow cytometry to provide highly quantitative analysis of mononuclear phagocytes in laser-induced choroidal neovascularization. Additionally, multi-parameter flow cytometry can be used for the identification of macrophage subsets, fate mapping, and cell sorting for transcriptomic or proteomic studies.


Subject(s)
Eye/cytology , Eye/diagnostic imaging , Flow Cytometry , Phagocytes/cytology , Animals , Antibodies/metabolism , Dendritic Cells/cytology , Female , Fluorescent Dyes/metabolism , Lasers , Macrophages/cytology , Male , Mice, Inbred C57BL , Microglia/cytology , Monocytes/cytology , Phagocytes/immunology
10.
Front Immunol ; 11: 230, 2020.
Article in English | MEDLINE | ID: mdl-32174913

ABSTRACT

Neuropsychiatric symptoms of systemic lupus erythematosus (NP-SLE) affect over one-half of SLE patients, yet underlying mechanisms remain largely unknown. We demonstrate that SLE-prone mice (CReCOM) develop NP-SLE, including behavioral deficits prior to systemic autoimmunity, reduced brain volumes, decreased vascular integrity, and brain-infiltrating leukocytes. NP-SLE microglia exhibit numerical expansion, increased synaptic uptake, and a more metabolically active phenotype. Microglia from multiple SLE-prone models express a "NP-SLE signature" unrelated to type I interferon. Rather, the signature is associated with lipid metabolism, scavenger receptor activity and downregulation of inflammatory and chemotaxis processes, suggesting a more regulatory, anti-inflammatory profile. NP-SLE microglia also express genes associated with disease-associated microglia (DAM), a subset of microglia thought to be instrumental in neurodegenerative diseases. Further, expression of "NP-SLE" and "DAM" signatures correlate with the severity of behavioral deficits in young SLE-prone mice prior to overt systemic disease. Our data are the first to demonstrate the predictive value of our newly identified microglia-specific "NP-SLE" and "DAM" signatures as a surrogate for NP-SLE clinical outcomes and suggests that microglia-intrinsic defects precede contributions from systemic SLE for neuropsychiatric manifestations.


Subject(s)
Lupus Erythematosus, Systemic/complications , Lupus Vasculitis, Central Nervous System/genetics , Memory Disorders/etiology , Microglia/metabolism , Transcriptome , Animals , Association Learning , Blood-Brain Barrier , Disease Models, Animal , Female , Genetic Predisposition to Disease , Gray Matter/diagnostic imaging , Gray Matter/pathology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Vasculitis, Central Nervous System/immunology , Lupus Vasculitis, Central Nervous System/pathology , Macrophages/metabolism , Maze Learning , Memory Disorders/genetics , Memory Disorders/immunology , Mice , Mice, Inbred MRL lpr , Mice, Mutant Strains , Morris Water Maze Test , Organ Size , Predictive Value of Tests , Prepulse Inhibition , Reflex, Startle , White Matter/diagnostic imaging , White Matter/pathology
11.
Invest Ophthalmol Vis Sci ; 60(15): 5059-5069, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31800964

ABSTRACT

Purpose: Beta-adrenergic receptor (AR) antagonists, like propranolol, inhibit angiogenesis in multiple ocular conditions through an unknown mechanism. We previously showed that propranolol reduces choroidal neovascularization (CNV) by decreasing interleukin-6 levels. Since macrophages are one of the central producers of interleukin-6, we examined whether macrophages are required for propranolol-driven inhibition of choroidal angiogenesis. Methods: We tested the anti-angiogenic properties of propranolol in the choroidal sprouting assay and the laser-induced CNV model. Bone marrow-derived monocytes (BMDMs) were added to the choroidal sprouting assay and Ccr2-/- mice were subjected to laser-induced CNV. Multi-parameter flow cytometry was performed to characterize the ocular mononuclear phagocyte populations after laser injury and during propranolol treatment. Results: Propranolol reduced choroidal angiogenesis by 41% (P < 0.001) in the choroidal sprouting assay. Similarly, propranolol decreased laser-induced CNV by 50% (P < 0.05) in female mice, with no change in males. BMDMs increased choroidal sprouting by 146% (P < 0.0001), and this effect was ablated by propranolol. Beta-AR inhibition had no effect upon laser-induced CNV area in female Ccr2-/- mice. MHCII+ and MHCII- macrophages increased 20-fold following laser treatment in wildtype mice as compared to untreated mice, and this effect was completely attenuated in lasered Ccr2-/- mice. Moreover, propranolol increased the numbers of MHCII+ and MHCII- macrophages by 1.9 (P = 0.07) and 3.1 (P < 0.05) fold in lasered female mice with no change in macrophage numbers in males. Conclusions: Our data suggest that propranolol inhibits angiogenesis through recruitment of monocyte-derived macrophages in female mice only. These data show the anti-angiogenic nature of beta-AR blocker-recruited monocyte-derived macrophages in CNV.


Subject(s)
Choroidal Neovascularization/drug therapy , Fluorescein Angiography/methods , Macrophages/pathology , Monocytes/pathology , Propranolol/pharmacology , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Choroid/metabolism , Choroid/pathology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Female , Flow Cytometry , Fundus Oculi , Imaging, Three-Dimensional , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Receptors, Adrenergic, beta/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
12.
Biochem Biophys Rep ; 14: 1-6, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29872727

ABSTRACT

αB-Crystallin, ubiquitously expressed in many tissues including the ocular lens, is a small heat shock protein that can prevent protein aggregation. A number of post-translation modifications are reported to modify αB-crystallin function. Recent studies have identified αB-crystallin lysine residues are modified by acetylation and ubiquitination. Therefore, we sought to determine the effects of lysine to alanine substitution on αB-crystallin functions including chaperone activity and modulation of actin polymerization. Analysis of the ten substitution mutants as recombinant proteins indicated all the proteins were soluble and formed oligomeric complexes similar to wildtype protein. Lysozyme aggregation induced by chemical treatment indicated that K82, K90, K121, K166 and K174/K175 were required for efficient chaperone activity. Thermal induction of γ-crystallin aggregation could be prevented by all αB-crystallin substitution mutants. These αB-crystallin mutants also were able to mediate wildtype levels of actin polymerization. Further analysis of two clones with either enhanced or reduced chaperone activity on individual client substrates or actin polymerization indicated both retained broad chaperone activity and anti-apoptotic activity. Collectively, these studies show the requirements for lysine residues in αB-crystallin function.

14.
Life Sci ; 91(21-22): 1070-6, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-22940618

ABSTRACT

AIM: To characterize how nicotinic acetylcholine receptors (nAChRs) influence epidermal barrier function and recovery following prolonged stress or direct nAChR activation or antagonism. MAIN METHODS: Mice were subjected to psychological stress or treated topically with nAChR agonist or antagonist for 3 days. We assessed barrier permeability and recovery by measuring transepidermal water loss (TEWL) before and after barrier disruption. In parallel, we analyzed the production and localization of several epidermal cornified envelope proteins in mouse skin and in human EpiDerm™ organotypic constructs stimulated with a nAChR agonist (nicotine) and/or a nAChR selective antagonist (α-bungarotoxin). KEY FINDINGS: We determined that psychological stress in mice impairs barrier permeability function and recovery, an effect that is reversed by application of the α7 selective nAChR antagonist, α-bungarotoxin (Bung). In the absence of stress, both topical nicotine or Bung treatment alone impaired barrier permeability. We further observed that stress, topical nicotine, or topical Bung treatment in mice influenced the abundance and/or localization of filaggrin, loricrin, and involucrin. Similar alterations in these three major cornified envelope proteins were observed in human EpiDerm™ cultures. SIGNIFICANCE: Perceived psychological stress and nicotine usage can both initiate or exacerbate several dermatoses by altering the cutaneous permeability barrier. Modulation of nAChRs by topical agonists or antagonists may be used to improve epidermal barrier function in skin diseases associated with defects in epidermal barrier permeability.


Subject(s)
Bungarotoxins/pharmacology , Cholinergic Antagonists/pharmacology , Epidermis/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Epidermis/physiology , Epidermis/ultrastructure , Filaggrin Proteins , Humans , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/metabolism , Male , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Permeability/drug effects , Protein Precursors/analysis , Protein Precursors/metabolism , Skin Absorption/drug effects , Stress, Physiological
15.
J Neurosci ; 32(23): 7895-906, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22674265

ABSTRACT

Neural networks called central pattern generators (CPGs) generate repetitive motor behaviors such as locomotion and breathing. Glutamatergic neurons are required for the generation and inhibitory neurons for the patterning of the motor activity associated with repetitive motor behaviors. In the mouse, glutamatergic V2a neurons coordinate the activity of left and right leg CPGs in the spinal cord enabling mice to generate an alternating gait. Here, we investigate the role of V2a neurons in the neural control of breathing, an essential repetitive motor behavior. We find that, following the ablation of V2a neurons, newborn mice breathe at a lower frequency. Recordings of respiratory activity in brainstem-spinal cord and respiratory slice preparations demonstrate that mice lacking V2a neurons are deficient in central respiratory rhythm generation. The absence of V2a neurons in the respiratory slice preparation can be compensated for by bath application of neurochemicals known to accelerate the breathing rhythm. In this slice preparation, V2a neurons exhibit a tonic firing pattern. The existence of direct connections between V2a neurons in the medial reticular formation and neurons of the pre-Bötzinger complex indicates that V2a neurons play a direct role in the function of the respiratory CPG in newborn mice. Thus, neurons of the embryonic V2a lineage appear to have been recruited to neural networks that control breathing and locomotion, two prominent CPG-driven, repetitive motor behaviors.


Subject(s)
Interneurons/physiology , Respiration/genetics , Animals , Animals, Newborn , Cell Count , Data Interpretation, Statistical , Electrophysiological Phenomena , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , In Situ Hybridization , Male , Medulla Oblongata/cytology , Medulla Oblongata/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Microscopy, Confocal , Microscopy, Video , Nerve Net/physiology , Plethysmography, Whole Body , Rhombencephalon/cytology , Rhombencephalon/physiology , Transcription Factors/genetics , Transcription Factors/physiology
16.
J Neurosci ; 30(1): 170-82, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053899

ABSTRACT

The V2a class of Chx10-expressing interneurons has been implicated in frequency-dependent control of left-right phase during locomotion in the mouse. We have used the Chx10::CFP mouse line to further investigate the properties and locomotion-related activity of V2a interneurons in the isolated neonatal spinal cord. V2a interneurons can be divided into three classes, based on their tonic, phasic, or delayed-onset responses to step depolarization. Electrical coupling is found only between neurons of same class and helps to synchronize neuronal activity within the class. Serotonin (5-HT) excites isolated tonic V2a interneurons by depolarizing the neurons and increasing their membrane input resistance, with no significant effects on action potential properties, a mechanism distinct from 5-HT excitation of commissural interneurons. During NMDA-/5-HT-induced locomotor-like activity, patch-clamp recordings and two-photon calcium imaging experiments show that approximately half of V2a interneurons fire rhythmically with ventral root-recorded motor activity; the rhythmic V2a interneurons fired during one half of the cycle, in phase with either the ipsilateral or the contralateral L2 ventral root bursts. The percentage of rhythmically firing V2a interneurons increases during higher-frequency fictive locomotion, and they become significantly more rhythmic in their firing during the locomotor cycle; this may help to explain the frequency-dependent shift in left-right coupling in Chx10::DTA mice, which lack these neurons. Our results together with data from the accompanying paper (Dougherty and Kiehn, 2009) reinforce earlier proposals that the V2a interneurons are components of the hindlimb central pattern generator, helping to organize left-right locomotor coordination in the neonatal mouse spinal cord.


Subject(s)
Interneurons/classification , Interneurons/physiology , Motor Activity/physiology , Spinal Cord/physiology , Animals , Animals, Newborn , Anterior Horn Cells/physiology , Electrophysiological Phenomena/physiology , Homeodomain Proteins/physiology , Membrane Potentials/physiology , Mice , Mice, Inbred ICR , Mice, Transgenic , Transcription Factors/physiology
17.
Neuron ; 60(1): 70-83, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18940589

ABSTRACT

The initiation and coordination of activity in limb muscles are the main functions of neural circuits that control locomotion. Commissural neurons connect locomotor circuits on the two sides of the spinal cord, and represent the known neural substrate for left-right coordination. Here we demonstrate that a group of ipsilateral interneurons, V2a interneurons, plays an essential role in the control of left-right alternation. In the absence of V2a interneurons, the spinal cord fails to exhibit consistent left-right alternation. Locomotor burst activity shows increased variability, but flexor-extensor coordination is unaffected. Anatomical tracing studies reveal a direct excitatory input of V2a interneurons onto commissural interneurons, including a set of molecularly defined V0 neurons that drive left-right alternation. Our findings imply that the neural substrate for left-right coordination consists of at least two components; commissural neurons and a class of ipsilateral interneurons that activate commissural pathways.


Subject(s)
Functional Laterality/physiology , Gene Deletion , Interneurons/physiology , Motor Activity/physiology , Recombination, Genetic , Spinal Cord/physiology , Afferent Pathways/physiology , Animals , Electric Stimulation/methods , Female , Functional Laterality/genetics , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Psychomotor Performance/physiology , Transcription Factors/deficiency , Transcription Factors/genetics
18.
Eur J Neurosci ; 26(11): 2989-3002, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18028107

ABSTRACT

The ventral spinal cord consists of interneuron groups arising from distinct, genetically defined, progenitor domains along the dorsoventral axis. Many of these interneuron groups settle in the ventral spinal cord which, in mammals, contains the central pattern generator for locomotion. In order to better understand the locomotor networks, we have used different transgenic mice for anatomical characterization of one of these interneuron groups, called V2 interneurons. Neurons in this group are either V2a interneurons marked by the postmitotic expression of the transcription factor Chx10, or V2b interneurons which express the transcription factors Gata2 and Gata3. We found that all V2a and most V2b interneurons were ipsilaterally projecting in embryos as well as in newborns. V2a interneurons were for the most part glutamatergic while V2b interneurons were mainly GABAergic or glycinergic. Furthermore, we demonstrated that a large proportion of V2 interneurons expressed the axon guidance molecule EphA4, a molecule previously shown to be important for correct organization of locomotor networks. We also showed that V2 interneurons and motor neurons alone did not account for all EphA4-expressing neurons in the spinal cord. Together, these findings enable a better interpretation of neural networks underlying locomotion, and open up the search for as yet unknown components of the mammalian central pattern generator.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Interneurons/physiology , Phenotype , Receptor, EphA4/metabolism , Spinal Cord , Animals , Animals, Newborn , Axons/physiology , Embryo, Mammalian , Functional Laterality , Glutamate Decarboxylase/genetics , Glycine Plasma Membrane Transport Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Interneurons/classification , Interneurons/cytology , LIM-Homeodomain Proteins , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/metabolism , Receptor, EphA4/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/growth & development , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 99(14): 9237-42, 2002 Jul 09.
Article in English | MEDLINE | ID: mdl-12077323

ABSTRACT

The function of GATA transcription factors in diverse developmental contexts depends in part on physical interaction with cofactors of the Friend of GATA (FOG) family. However, previous studies indicate that FOG-1 may play a GATA-1-independent role in early megakaryopoiesis, suggesting that FOG proteins might act in a GATA factor-independent manner. Here, we have generated mouse knock-in (KI) mutants harboring a critical valine-to-glycine substitution in the amino-terminal zinc fingers of GATA-1 and GATA-2 to ablate FOG interaction. In contrast to male GATA-1(KI) (GATA-1 is located on the X-chromosome) or GATA-2(KI/KI) mice, compound GATA-1(KI) GATA-2(KI/KI) mutant mice display complete megakaryopoietic failure, a phenocopy of FOG-1(-/-) mice. We conclude that FOG-1 requires an interaction with either GATA-1 or -2 as part of its essential role in early megakaryopoiesis. On the basis of these and previous reports, we infer that GATA factor dependence is a critical aspect of FOG protein function.


Subject(s)
Carrier Proteins/physiology , DNA-Binding Proteins/physiology , Hematopoiesis/physiology , Megakaryocytes/cytology , Megakaryocytes/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Erythroid-Specific DNA-Binding Factors , Female , GATA1 Transcription Factor , GATA2 Transcription Factor , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Molecular Sequence Data , Mutagenesis, Site-Directed , Transcription Factors/chemistry , Transcription Factors/genetics , Zinc Fingers/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...