Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Vision Res ; 214: 108339, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38039846

ABSTRACT

Retinal function changes dramatically from day to night, yet clinical diagnosis, treatments, and experimental sampling occur during the day. To begin to address this gap in our understanding of disease pathobiology, this study investigates whether diabetes affects the retina's daily rhythm of gene expression. Diabetic, Ins2Akita/J mice, and non-diabetic littermates were kept under a 12 h:12 h light/dark cycle until 4 months of age. mRNA sequencing was conducted in retinas collected every 4 h throughout the 24 hr light/dark cycle. Computational approaches were used to detect rhythmicity, predict acrophase, identify differential rhythmic patterns, analyze phase set enrichment, and predict upstream regulators. The retinal transcriptome exhibited a tightly regulated rhythmic expression with a clear 12-hr transcriptional axis. Day-peaking genes were enriched for DNA repair, RNA splicing, and ribosomal protein synthesis, night-peaking genes for metabolic processes and growth factor signaling. Although the 12-hr transcriptional axis is retained in the diabetic retina, it is phase advanced for some genes. Upstream regulator analysis for the phase-shifted genes identified oxygen-sensing mechanisms and HIF1alpha, but not the circadian clock, which remained in phase with the light/dark cycle. We propose a model in which, early in diabetes, the retina is subjected to an internal desynchrony with the circadian clock and its outputs are still light-entrained whereas metabolic pathways related to neuronal dysfunction and hypoxia are phase advanced. Further studies are now required to evaluate the chronic implications of such desynchronization on the development of diabetic retinopathy.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Mice , Animals , Circadian Rhythm/genetics , Transcriptome , Retina/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Photoperiod
2.
Diabetologia ; 64(8): 1822-1833, 2021 08.
Article in English | MEDLINE | ID: mdl-34003304

ABSTRACT

AIMS/HYPOTHESIS: The circadian clock influences both diabetes and immunity. Our goal in this study was to characterise more thoroughly the circadian patterns of immune cell populations and cytokines that are particularly relevant to the immune pathology of type 1 diabetes and thus fill in a current gap in our understanding of this disease. METHODS: Ten individuals with established type 1 diabetes (mean disease duration 11 years, age 18-40 years, six female) participated in a circadian sampling protocol, each providing six blood samples over a 24 h period. RESULTS: Daily ranges of population frequencies were sometimes large and possibly clinically significant. Several immune populations, such as dendritic cells, CD4 and CD8 T cells and their effector memory subpopulations, CD4 regulatory T cells, B cells and cytokine IL-6, exhibited statistically significant circadian rhythmicity. In a comparison with historical healthy control individuals, but using shipped samples, we observed that participants with type 1 diabetes had statistically significant phase shifts occurring in the time of peak occurrence of B cells (+4.8 h), CD4 and CD8 T cells (~ +5 h) and their naive and effector memory subsets (~ +3.3 to +4.5 h), and regulatory T cells (+4.1 h). An independent streptozotocin murine experiment confirmed the phase shifting of CD8 T cells and suggests that circadian dysrhythmia in type 1 diabetes might be an effect and not a cause of the disease. CONCLUSIONS/INTERPRETATION: Future efforts investigating this newly described aspect of type 1 diabetes in human participants are warranted. Peripheral immune populations should be measured near the same time of day in order to reduce circadian-related variation.


Subject(s)
Chronobiology Disorders/immunology , Circadian Rhythm/immunology , Diabetes Mellitus, Type 1/immunology , Immune System/physiology , Adolescent , Adult , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Circadian Clocks/genetics , Dendritic Cells/immunology , Female , Flow Cytometry , Humans , Interleukin-6/blood , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Young Adult
3.
Mil Med ; 185(Suppl 1): 404-412, 2020 01 07.
Article in English | MEDLINE | ID: mdl-32074302

ABSTRACT

INTRODUCTION: Whole-body vibration training (WBVT) may benefit individuals with difficulty participating in physical exercise. The objective was to explore the effects of WBVT on circulating stem/progenitor cell (CPC) and cytokine levels. METHODS: Healthy male subjects each performed three activities randomly on separate days: (1) standing platform vibration, (2) repetitive leg squat exercise; and (3) in combination. Pre- and post-activity blood samples were drawn. Cell populations were characterized using flow cytometry. Biomarkers were analyzed using enzyme-linked immunosorbent assays. RESULTS: CPC levels increased significantly 21% with exercise alone (1465 ± 202-1770 ± 221 cells/mL; P = 0.017) and 33% with vibration alone in younger participants (1918 ± 341-2559 ± 496; P = 0.02). Angiogenic CPCs increased 39% during combined activity in younger (633 ± 128-882 ± 181; P = 0.05). Non-angiogenic CPCs increased 42% with vibration alone in younger (1181 ± 222-1677 ± 342; P = 0.04), but 32% with exercise alone in older participants (801 ± 251-1053 ± 325; P = 0.05). With vibration alone, anti-inflammatory cytokine interleukin-10 increased significantly (P < 0.03), although inflammatory interleukin-6 decreased (P = 0.056); tumor necrosis factor-alpha (P < 0.01) and vascular endothelial growth factor levels increased (P < 0.005), which are synergistically pro-angiogenic. CONCLUSIONS: WBVT may have positive vascular and anti-inflammatory effects. WBVT could augment or serve as an exercise surrogate in warfighters and others who cannot fully participate in exercise programs, having important implications in military health.


Subject(s)
Inflammation/therapy , Physical Therapy Modalities/trends , Stem Cells/physiology , Teaching/statistics & numerical data , Vibration/therapeutic use , Adult , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Male , Stem Cells/metabolism
4.
Circ Res ; 125(11): 969-988, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31610731

ABSTRACT

RATIONALE: There is incomplete knowledge of the impact of bone marrow cells on the gut microbiome and gut barrier function. OBJECTIVE: We postulated that diabetes mellitus and systemic ACE2 (angiotensin-converting enzyme 2) deficiency would synergize to adversely impact both the microbiome and gut barrier function. METHODS AND RESULTS: Bacterial 16S rRNA sequencing and metatranscriptomic analysis were performed on fecal samples from wild-type, ACE2-/y, Akita (type 1 diabetes mellitus), and ACE2-/y-Akita mice. Gut barrier integrity was assessed by immunofluorescence, and bone marrow cell extravasation into the small intestine was evaluated by flow cytometry. In the ACE2-/y-Akita or Akita mice, the disrupted barrier was associated with reduced levels of myeloid angiogenic cells, but no increase in inflammatory monocytes was observed within the gut parenchyma. Genomic and metatranscriptomic analysis of the microbiome of ACE2-/y-Akita mice demonstrated a marked increase in peptidoglycan-producing bacteria. When compared with control cohorts treated with saline, intraperitoneal administration of myeloid angiogenic cells significantly decreased the microbiome gene expression associated with peptidoglycan biosynthesis and restored epithelial and endothelial gut barrier integrity. Also indicative of diabetic gut barrier dysfunction, increased levels of peptidoglycan and FABP-2 (intestinal fatty acid-binding protein 2) were observed in plasma of human subjects with type 1 diabetes mellitus (n=21) and type 2 diabetes mellitus (n=23) compared with nondiabetic controls (n=23). Using human retinal endothelial cells, we determined that peptidoglycan activates a noncanonical TLR-2 (Toll-like receptor 2) associated MyD88 (myeloid differentiation primary response protein 88)-ARNO (ADP-ribosylation factor nucleotide-binding site opener)-ARF6 (ADP-ribosylation factor 6) signaling cascade, resulting in destabilization of p120-catenin and internalization of VE-cadherin as a mechanism of deleterious impact of peptidoglycan on the endothelium. CONCLUSIONS: We demonstrate for the first time that the defect in gut barrier function and dysbiosis in ACE2-/y-Akita mice can be favorably impacted by exogenous administration of myeloid angiogenic cells.


Subject(s)
Bacteria/metabolism , Bone Marrow Transplantation , Capillary Permeability , Diabetes Mellitus, Type 2/surgery , Gastrointestinal Microbiome , Intestinal Mucosa/blood supply , Intestinal Mucosa/microbiology , Intestine, Small/blood supply , Intestine, Small/microbiology , Neovascularization, Physiologic , Peptidyl-Dipeptidase A/deficiency , ADP-Ribosylation Factor 6 , Adherens Junctions/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Dysbiosis , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/enzymology , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Peptidoglycan/metabolism , Peptidyl-Dipeptidase A/genetics , Recovery of Function
5.
Nutrients ; 11(10)2019 Sep 29.
Article in English | MEDLINE | ID: mdl-31569518

ABSTRACT

Our hypothesis is that diabetes leads to loss of diurnal oscillatory rhythms in gut microbiota altering circulating metabolites. We performed an observational study where we compared diurnal changes of the gut microbiota with temporal changes of plasma metabolites. Metadata analysis from bacterial DNA from fecal pellets collected from 10-month old control (db/m) and type 2 diabetic (db/db) mice every 4 h for a 24-h period was used for prediction analysis. Blood plasma was collected at a day and night time points and was used for untargeted global metabolomic analysis. Feeding and activity behaviors were recorded. Our results show that while diabetic mice exhibited feeding and activity behavior similar to control mice, they exhibited a loss of diurnal oscillations in bacteria of the genus Akkermansia, Bifidobacterium, Allobaculum, Oscillospira and a phase shift in the oscillations of g.Prevotella, proteobacteria, and actinobacteria. Analysis of the circulating metabolites showed alterations in the diurnal pattern of metabolic pathways where bacteria have been implicated, such as the histidine, betaine, and methionine/cysteine pathway, mitochondrial function and the urea cycle. Functional analysis of the differential microbes revealed that during the day, when mice are asleep, the microbes of diabetic mice were enriched in processing carbon and pyruvate metabolic pathways instead of xenobiotic degradation as was observed for control mice. Altogether, our study suggests that diabetes led to loss of rhythmic oscillations of many gut microbiota with possible implications for temporal regulation of host metabolic pathways.


Subject(s)
Circadian Rhythm/physiology , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/physiology , Metabolome/physiology , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Feces/microbiology , Mice
6.
Diabetes ; 67(9): 1867-1879, 2018 09.
Article in English | MEDLINE | ID: mdl-29712667

ABSTRACT

Intermittent fasting (IF) protects against the development of metabolic diseases and cancer, but whether it can prevent diabetic microvascular complications is not known. In db/db mice, we examined the impact of long-term IF on diabetic retinopathy (DR). Despite no change in glycated hemoglobin, db/db mice on the IF regimen displayed significantly longer survival and a reduction in DR end points, including acellular capillaries and leukocyte infiltration. We hypothesized that IF-mediated changes in the gut microbiota would produce beneficial metabolites and prevent the development of DR. Microbiome analysis revealed increased levels of Firmicutes and decreased Bacteroidetes and Verrucomicrobia. Compared with db/db mice on ad libitum feeding, changes in the microbiome of the db/db mice on IF were associated with increases in gut mucin, goblet cell number, villi length, and reductions in plasma peptidoglycan. Consistent with the known modulatory effects of Firmicutes on bile acid (BA) metabolism, measurement of BAs demonstrated a significant increase of tauroursodeoxycholate (TUDCA), a neuroprotective BA, in db/db on IF but not in db/db on AL feeding. TGR5, the TUDCA receptor, was found in the retinal primary ganglion cells. Expression of TGR5 did not change with IF or diabetes. However, IF reduced retinal TNF-α mRNA, which is a downstream target of TGR5 activation. Pharmacological activation of TGR5 using INT-767 prevented DR in a second diabetic mouse model. These findings support the concept that IF prevents DR by restructuring the microbiota toward species producing TUDCA and subsequent retinal protection by TGR5 activation.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Diabetic Retinopathy/prevention & control , Dysbiosis/therapy , Fasting , Gastrointestinal Microbiome , Retina/pathology , Retinal Vessels/pathology , Animals , Bacteroidetes/growth & development , Bacteroidetes/immunology , Bacteroidetes/isolation & purification , Bile Acids and Salts/therapeutic use , Colon/drug effects , Colon/immunology , Colon/metabolism , Colon/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/pathology , Diabetic Retinopathy/complications , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Dysbiosis/complications , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Firmicutes/growth & development , Firmicutes/immunology , Firmicutes/isolation & purification , Ganglia, Sensory/drug effects , Ganglia, Sensory/immunology , Ganglia, Sensory/metabolism , Ganglia, Sensory/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Goblet Cells/drug effects , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/pathology , Male , Mice, Inbred DBA , Mice, Mutant Strains , Microvessels/drug effects , Microvessels/immunology , Microvessels/metabolism , Microvessels/pathology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Retina/drug effects , Retina/immunology
7.
Stem Cells ; 36(9): 1430-1440, 2018 09.
Article in English | MEDLINE | ID: mdl-29761600

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is the primary enzyme of the vasoprotective axis of the renin angiotensin system (RAS). We tested the hypothesis that loss of ACE2 would exacerbate diabetic retinopathy by promoting bone marrow dysfunction. ACE2-/y were crossed with Akita mice, a model of type 1 diabetes. When comparing the bone marrow of the ACE2-/y -Akita mice to that of Akita mice, we observed a reduction of both short-term and long-term repopulating hematopoietic stem cells, a shift of hematopoiesis toward myelopoiesis, and an impairment of lineage- c-kit+ hematopoietic stem/progenitor cell (HS/PC) migration and proliferation. Migratory and proliferative dysfunction of these cells was corrected by exposure to angiotensin-1-7 (Ang-1-7), the protective peptide generated by ACE2. Over the duration of diabetes examined, ACE2 deficiency led to progressive reduction in electrical responses assessed by electroretinography and to increases in neural infarcts observed by fundus photography. Compared with Akita mice, ACE2-/y -Akita at 9-months of diabetes showed an increased number of acellular capillaries indicative of more severe diabetic retinopathy. In diabetic and control human subjects, CD34+ cells, a key bone marrow HS/PC population, were assessed for changes in mRNA levels for MAS, the receptor for Ang-1-7. Levels were highest in CD34+ cells from diabetics without retinopathy. Higher serum Ang-1-7 levels predicted protection from development of retinopathy in diabetics. Treatment with Ang-1-7 or alamandine restored the impaired migration function of CD34+ cells from subjects with retinopathy. These data support that activation of the protective RAS within HS/PCs may represents a therapeutic strategy for prevention of diabetic retinopathy. Stem Cells 2018;36:1430-1440.


Subject(s)
Bone Marrow/metabolism , Diabetic Retinopathy/chemically induced , Peptidyl-Dipeptidase A/adverse effects , Peptidyl-Dipeptidase A/deficiency , Angiotensin-Converting Enzyme 2 , Animals , Disease Models, Animal , Humans , Mice
8.
Invest Ophthalmol Vis Sci ; 58(12): 5164-5176, 2017 10 01.
Article in English | MEDLINE | ID: mdl-29049716

ABSTRACT

Purpose: We investigated whether subthreshold retinal phototherapy (SRPT) was associated with recruitment of bone marrow (BM)-derived cells to the neurosensory retina (NSR) and RPE layer. Methods: GFP chimeric mice and wild-type (WT) mice were subjected to SRPT using a slit-lamp infrared laser. Duty cycles of 5%, 10%, 15%, and 20% (0.1 seconds, 250 mW, spot size 50 µm) with 30 applications were placed 50 to 100 µm from the optic disc. In adoptive transfer studies, GFP+ cells were given intravenously immediately after WT mice received SRPT. Immunohistochemistry was done for ionized calcium-binding adapter molecule-1 (IBA-1+), CD45, Griffonia simplicifolia lectin isolectin B4, GFP or cytokeratin). Expression of Ccl2, Il1b, Il6, Hspa1a, Hsp90aa1, Cryab, Hif1a, Cxcl12, and Cxcr4 mRNA and flow cytometry of the NSR and RPE-choroid were performed. Results: Within 12 to 24 hours of SRPT, monocytes were detected in the NSR and RPE-choroid. Detection of reparative progenitors in the RPE occurred at 2 weeks using flow cytometry. Recruitment of GFP+ cells to the RPE layer occurred in a duty cycle-dependent manner in chimeric mice and in mice undergoing adoptive transfer. Hspa1a, Hsp90aa1, and Cryab mRNAs increased in the NSR at 2 hours post laser; Hif1a, Cxcl12, Hspa1a increased at 4 hours in the RPE-choroid; and Ccl2, Il1b, Ifng, and Il6 increased at 12 to 24 hours in the RPE-choroid. Conclusions: SRPT induces monocyte recruitment to the RPE followed by hematopoietic progenitor cell homing at 2 weeks. Recruitment occurs in a duty cycle-dependent manner and potentially could contribute to the therapeutic efficacy of SRPT.


Subject(s)
Bone Marrow Cells/physiology , Cell Movement/physiology , Phototherapy , Retina/cytology , Retinal Pigment Epithelium/cytology , Adoptive Transfer , Animals , Biomarkers/metabolism , Cells, Cultured , Chemokine CXCL12/metabolism , Choroid/cytology , Choroid/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Immunohistochemistry , Laser Therapy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/physiology , Receptors, CXCR4/metabolism , Retina/metabolism , Retina/surgery , Retinal Pigment Epithelium/metabolism
9.
EBioMedicine ; 22: 181-190, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28774737

ABSTRACT

Diabetic retinopathy (DR) is a complication secondary to diabetes and is the number one cause of blindness among working age individuals worldwide. Despite recent therapeutic breakthroughs using pharmacotherapy, a cure for DR has yet to be realized. Several clinical trials have highlighted the vital role dyslipidemia plays in the progression of DR. Additionally, it has recently been shown that activation of Liver X receptor (LXRα/LXRß) prevents DR in diabetic animal models. LXRs are nuclear receptors that play key roles in regulating cholesterol metabolism, fatty acid metabolism and inflammation. In this manuscript, we show insight into DR pathogenesis by demonstrating an innovative signaling axis that unifies key metabolic regulators, Sirtuin 1 and LXR, in modulating retinal cholesterol metabolism and inflammation in the diabetic retina. Expression of both regulators, Sirtuin 1 and LXR, are significantly decreased in diabetic human retinal samples and in a type 2 diabetic animal model. Additionally, activation of LXR restores reverse cholesterol transport, prevents inflammation, reduces pro-inflammatory macrophages activity and prevents the formation of diabetes-induced acellular capillaries. Taken together, the work presented in this manuscript highlights the important role lipid dysregulation plays in DR progression and offers a novel potential therapeutic target for the treatment of DR.


Subject(s)
Cholesterol/metabolism , Diabetic Retinopathy/metabolism , Liver X Receptors/metabolism , Sirtuin 1/metabolism , Animals , Cattle , Cells, Cultured , Disease Models, Animal , Down-Regulation , Humans , Mice , Retina/metabolism , Signal Transduction
10.
Basic Res Cardiol ; 112(5): 54, 2017 09.
Article in English | MEDLINE | ID: mdl-28756533

ABSTRACT

Coronary transient receptor potential canonical (TRPC) channel expression is elevated in metabolic syndrome (MetS). However, differential contribution of TRPCs to coronary pathology in MetS is not fully elucidated. We investigated the roles of TRPC1 and TRPC6 isoforms in coronary arteries of MetS pigs and determined whether long-term treatment with a mineralocorticoid receptor inhibitor, spironolactone, attenuates coronary TRPC expression and associated dysfunctions. MetS coronary arteries exhibited significant atherosclerosis, endothelial dysfunction, and increased histamine-induced contractions. Immunohistochemical studies revealed that TRPC6 immunostaining was significantly greater in the medial layer of MetS pig coronary arteries compared to that in Lean pigs, whereas little TRPC6 immunostaining was found in atheromas. Conversely, TRPC1 immunostaining was weak in the medial layer but strong in MetS atheromas, where it was predominantly localized to macrophages. Spironolactone treatment significantly decreased coronary TRPC expression and dysfunctions in MetS pigs. In vivo targeted delivery of the dominant-negative (DN)-TRPC6 cDNA to the coronary wall reduced histamine-induced calcium transients in the MetS coronary artery medial layer, implying a role for TRPC6 in mediating calcium influx in MetS coronary smooth muscles. Monocyte adhesion was increased in Lean pig coronary arteries cultured in the presence of aldosterone; and spironolactone antagonized this effect, suggesting that coronary mineralocorticoid receptor activation may regulate macrophage infiltration. TRPC1 expression in atheroma macrophages was associated with advanced atherosclerosis, whereas medial TRPC6 upregulation correlated with increased histamine-induced calcium transients and coronary contractility. We propose that long-term spironolactone treatment may be a therapeutic strategy to decrease TRPC expression and coronary pathology associated with MetS.


Subject(s)
Coronary Artery Disease/prevention & control , Coronary Vessels/drug effects , Metabolic Syndrome/drug therapy , Mineralocorticoid Receptor Antagonists/administration & dosage , Spironolactone/administration & dosage , TRPC Cation Channels/drug effects , TRPC6 Cation Channel/drug effects , Vasoconstriction/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Disease Models, Animal , Down-Regulation , Drug Administration Schedule , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Macrophages/drug effects , Macrophages/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Swine , Swine, Miniature , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism , Time Factors , Tissue Culture Techniques
11.
J Clin Lipidol ; 11(3): 694-703, 2017.
Article in English | MEDLINE | ID: mdl-28457994

ABSTRACT

BACKGROUND: Diabetic retinopathy is a microvascular disease that results from retinal vascular degeneration and defective repair due to diabetes-induced endothelial progenitor dysfunction. OBJECTIVE: Understanding key molecular factors involved in vascular degeneration and repair is paramount for developing effective diabetic retinopathy treatment strategies. We propose that diabetes-induced activation of acid sphingomyelinase (ASM) plays essential role in retinal endothelial and CD34+ circulating angiogenic cell (CAC) dysfunction in diabetes. METHODS: Human retinal endothelial cells (HRECs) isolated from control and diabetic donor tissue and human CD34+ CACs from control and diabetic patients were used in this study. ASM messenger RNA and protein expression were assessed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. To evaluate the effect of diabetes-induced ASM on HRECs and CD34+ CACs function, tube formation, CAC incorporation into endothelial tubes, and diurnal release of CD34+ CACs in diabetic individuals were determined. RESULTS: ASM expression level was significantly increased in HRECs isolated from diabetic compared with control donor tissue, as well as CD34+ CACs and plasma of diabetic patients. A significant decrease in tube area was observed in HRECs from diabetic donors compared with control HRECs. The tube formation deficiency was associated with increased expression of ASM in diabetic HRECs. Moreover, diabetic CD34+ CACs with high ASM showed defective incorporation into endothelial tubes. Diurnal release of CD34+ CACs was disrupted with the rhythmicity lost in diabetic patients. CONCLUSION: Collectively, these findings support that diabetes-induced ASM upregulation has a marked detrimental effect on both retinal endothelial cells and CACs.


Subject(s)
Diabetic Retinopathy/enzymology , Endothelial Cells/metabolism , Neovascularization, Pathologic/pathology , Retina/pathology , Retinal Vessels/physiopathology , Sphingomyelin Phosphodiesterase/metabolism , Aged , Antigens, CD34/metabolism , Circadian Rhythm , Diabetic Retinopathy/blood , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Endothelial Cells/pathology , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Middle Aged , Retinal Vessels/metabolism , Sphingomyelin Phosphodiesterase/genetics
13.
Am J Pathol ; 187(6): 1426-1435, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28432873

ABSTRACT

The brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein (BMAL)-1 constitutes a major transcriptional regulator of the circadian clock. Here, we explored the impact of conditional deletion of Bmal1 in endothelium and hematopoietic cells in murine models of microvascular and macrovascular injury. We used two models of Bmal1fx/fx;Tek-Cre mice, a retinal ischemia/reperfusion model and a neointimal hyperplasia model of the femoral artery. Eyes were enumerated for acellular capillaries and were stained for oxidative damage markers using nitrotyrosine immunohistochemistry. LSK (lineage-negative, stem cell antigen-1-positive, c-Kit-positive) cells were quantified and proliferation assessed. Hematopoiesis is influenced by innervation to the bone marrow, which we assessed using IHC analysis. The number of acellular capillaries increased threefold, and nitrotyrosine staining increased 1.5-fold, in the retinas of Bmal1fx/fx;Tek-Cre mice. The number of LSK cells from the Bmal1fx/fx;Tek-Cre mice decreased by 1.5-fold and was accompanied by a profound decrease in proliferative potential. Bmal1fx/fx;Tek-Cre mice also exhibited evidence of bone marrow denervation, demonstrating a loss of neurofilament-200 staining. Injured femoral arteries showed a 20% increase in neointimal hyperplasia compared with similarly injured wild-type controls. Our study highlights the importance of the circadian clock in maintaining vascular homeostasis and demonstrates that specific deletion of BMAL1 in endothelial and hematopoietic cells results in phenotypic features similar to those of diabetes.


Subject(s)
ARNTL Transcription Factors/physiology , Neointima/pathology , Reperfusion Injury/metabolism , Retinal Vessels/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Capillaries/pathology , Cell Proliferation , Circadian Rhythm/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Femoral Artery/injuries , Femoral Artery/pathology , Gene Deletion , Hematopoietic Stem Cells/pathology , Hyperplasia , Leukocyte Common Antigens/analysis , Leukocyte Count , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/pathology , Retina/metabolism , Retinal Vessels/pathology
14.
Stem Cells ; 35(5): 1303-1315, 2017 05.
Article in English | MEDLINE | ID: mdl-28299842

ABSTRACT

Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.


Subject(s)
Central Nervous System/cytology , Electroacupuncture , Mesenchymal Stem Cells/cytology , Achilles Tendon/pathology , Acupuncture Points , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Antigens, CD/metabolism , Forelimb/physiology , Hindlimb/physiology , Humans , Hyperalgesia/therapy , Hypothalamus/cytology , Interleukin-10/blood , Macrophages/cytology , Mice , Nerve Net/physiology , Rats , Rupture , Sensory Receptor Cells/metabolism , Uncoupling Protein 1/metabolism
15.
PLoS One ; 12(2): e0171940, 2017.
Article in English | MEDLINE | ID: mdl-28222108

ABSTRACT

p62 is a scaffolding adaptor implicated in the clearance of protein aggregates by autophagy. Reactive oxygen species (ROS) can either stimulate or inhibit NFκB-mediated gene expression influencing cellular fate. We studied the effect of hydrogen peroxide (H2O2)-mediated oxidative stress and NFκB signaling on p62 expression in the retinal pigment epithelium (RPE) and investigated its role in regulation of autophagy and RPE survival against oxidative damage. Cultured human RPE cell line ARPE-19 and primary human adult and fetal RPE cells were exposed to H2O2-induced oxidative stress. The human apolipoprotein E4 targeted-replacement (APOE4) mouse model of AMD was used to study expression of p62 and other autophagy proteins in the retina. p62, NFκB p65 (total, phosphorylated, nuclear and cytoplasmic) and ATG10 expression was assessed by mRNA and protein analyses. Cellular ROS and mitochondrial superoxide were measured by CM-H2DCFDA and MitoSOX staining respectively. Mitochondrial viability was determined using MTT activity. qPCR-array system was used to investigate autophagic genes affected by p62. Nuclear and cytoplasmic levels of NFκB p65 were evaluated after cellular fractionation by Western blotting. We report that p62 is up-regulated in RPE cells under H2O2-induced oxidative stress and promotes autophagic activity. Depletion of endogenous p62 reduces autophagy by downregulation of ATG10 rendering RPE more susceptible to oxidative damage. NFκB p65 phosphorylation at Ser-536 was found to be critical for p62 upregulation in response to oxidative stress. Proteasome inhibition by H2O2 causes p62-NFκB signaling as antioxidant pre-treatment reversed p62 expression and p65 phosphorylation when RPE was challenged by H2O2 but not when by Lactacystin. p62 protein but not RNA levels are elevated in APOE4-HFC AMD mouse model, suggesting reduction of autophagic flux in disease conditions. Our findings suggest that p62 is necessary for RPE cytoprotection under oxidative stress and functions, in part, by modulating ATG10 expression. NFκB p65 activity may be a critical upstream initiator of p62 expression in RPE cells under oxidative stress.


Subject(s)
Autophagy/physiology , Cell Survival/physiology , NF-kappa B/physiology , Oxidative Stress/physiology , RNA-Binding Proteins/physiology , Retinal Pigment Epithelium/physiology , Sequestosome-1 Protein/physiology , Animals , Blotting, Western , Cell Line , Disease Models, Animal , Fluorescent Antibody Technique , Macular Degeneration/etiology , Macular Degeneration/physiopathology , Mice , Phosphorylation , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Superoxides/metabolism , Up-Regulation
16.
J Mol Med (Berl) ; 94(11): 1255-1265, 2016 11.
Article in English | MEDLINE | ID: mdl-27344677

ABSTRACT

In this study, the role of CX3CR1 in the progression of diabetic retinopathy (DR) was investigated. The retinas of wild-type (WT), CX3CR1 null (CX3CR1gfp/gfp, KO), and heterozygous (CX3CR1+/gfp, Het) mice were compared in the presence and absence of streptozotocin (STZ)-induced diabetes. CX3CR1 deficiency in STZ-KO increased vascular pathology at 4 months of diabetes, as a significant increase in acellular capillaries was observed only in the STZ-KO group. CX3CR1 deficiency and diabetes had similar effects on retinal neurodegeneration measured by an increase in DNA fragmentation. Retinal vascular pathology in STZ-KO mice was associated with increased numbers of monocyte-derived macrophages in the retina. Furthermore, compared to STZ-WT, STZ-KO mice exhibited increased numbers of inflammatory monocytes in the bone marrow and impaired homing of monocytes to the spleen. The induction of retinal IL-10 expression by diabetes was significantly less in KO mice, and when bone marrow-derived macrophages from KO mice were maintained in high glucose, they expressed significantly less IL-10 and more TNF-α in response to LPS stimulation. These findings support that CX3CR1 deficiency accelerates the development of vascular pathology in DR through increased recruitment of proinflammatory myeloid cells that demonstrate reduced expression of anti-inflammatory IL-10. KEY MESSAGES: • CX3CR1 deletion in STZ-diabetic mice accelerated the onset of diabetic retinopathy (DR). • The early onset of DR was associated with increased retinal cell apoptosis. • The early onset of DR was associated with increased recruitment of bone marrow-derived macrophages to the retina. • Bone marrow-derived macrophages from CX3CR1 KO diabetic mice expressed more TNF-α and less IL-10. • The role of IL-10 in protection from progression of DR is highlighted.


Subject(s)
CX3C Chemokine Receptor 1/deficiency , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Animals , Apoptosis , Body Weight , Bone Marrow Cells/metabolism , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Gene Deletion , Glycated Hemoglobin/metabolism , Homeostasis , Hypothalamus/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myeloid Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Streptozocin
17.
PLoS One ; 11(1): e0146829, 2016.
Article in English | MEDLINE | ID: mdl-26760976

ABSTRACT

Diabetic retinopathy is a sight-threatening complication of diabetes, affecting 65% of patients after 10 years of the disease. Diabetic metabolic insult leads to chronic low-grade inflammation, retinal endothelial cell loss and inadequate vascular repair. This is partly due to bone marrow (BM) pathology leading to increased activity of BM-derived pro-inflammatory monocytes and impaired function of BM-derived reparative circulating angiogenic cells (CACs). We propose that diabetes has a significant long-term effect on the nature and proportion of BM-derived cells that circulate in the blood, localize to the retina and home back to their BM niche. Using a streptozotocin mouse model of diabetic retinopathy with GFP BM-transplantation, we have demonstrated that BM-derived circulating pro-inflammatory monocytes are increased in diabetes while reparative CACs are trapped in the BM and spleen, with impaired release into circulation. Diabetes also alters activation of splenocytes and BM-derived dendritic cells in response to LPS stimulation. A majority of the BM-derived GFP cells that migrate to the retina express microglial markers, while others express endothelial, pericyte and Müller cell markers. Diabetes significantly increases infiltration of BM-derived microglia in an activated state, while reducing infiltration of BM-derived endothelial progenitor cells in the retina. Further, control CACs injected into the vitreous are very efficient at migrating back to their BM niche, whereas diabetic CACs have lost this ability, indicating that the in vivo homing efficiency of diabetic CACs is dramatically decreased. Moreover, diabetes causes a significant reduction in expression of specific integrins regulating CAC migration. Collectively, these findings indicate that BM pathology in diabetes could play a role in both increased pro-inflammatory state and inadequate vascular repair contributing to diabetic retinopathy.


Subject(s)
Bone Marrow Cells/cytology , Diabetic Retinopathy/pathology , Endothelial Cells/pathology , Inflammation/pathology , Animals , Bone Marrow Transplantation , Cell Count , Chimera , Dendritic Cells/pathology , Diabetic Retinopathy/immunology , Green Fluorescent Proteins/metabolism , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Microglia/pathology , Microvessels/drug effects , Microvessels/pathology , Monocytes/pathology , Retina/pathology , Spleen/pathology
18.
Stem Cells ; 34(4): 972-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26676316

ABSTRACT

The metabolic insults associated with diabetes lead to low-grade chronic inflammation, retinal endothelial cell damage, and inadequate vascular repair. This is partly due to the increased activation of bone marrow (BM)-derived proinflammatory monocytes infiltrating the retina, and the compromised function of BM-derived reparative circulating angiogenic cells (CACs), which home to sites of endothelial injury and foster vascular repair. We now propose that a metabolic link leading to activated monocytes and dysfunctional CACs in diabetes involves upregulation of a central enzyme of sphingolipid signaling, acid sphingomyelinase (ASM). Selective inhibition of ASM in the BM prevented diabetes-induced activation of BM-derived microglia-like cells and normalized proinflammatory cytokine levels in the retina. ASM upregulation in diabetic CACs caused accumulation of ceramide on their cell membrane, thereby reducing membrane fluidity and impairing CAC migration. Replacing sphingomyelin with ceramide in synthetic membrane vesicles caused a similar decrease in membrane fluidity. Inhibition of ASM in diabetic CACs improved membrane fluidity and homing of these cells to damaged retinal vessels. Collectively, these findings indicate that selective modulation of sphingolipid metabolism in BM-derived cell populations in diabetes normalizes the reparative/proinflammatory cell balance and can be explored as a novel therapeutic strategy for treating diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/genetics , Diabetic Retinopathy/therapy , Retina/growth & development , Retinal Vessels/metabolism , Sphingomyelin Phosphodiesterase/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Ceramides/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/therapy , Mice , Monocytes/metabolism , Monocytes/pathology , Retina/metabolism , Retina/pathology , Retinal Vessels/growth & development , Retinal Vessels/pathology , Sphingolipids/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism
19.
Mech Ageing Dev ; 135: 33-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24361677

ABSTRACT

The effect of aging on natural killer cell homeostasis is not well studied in humans or in animal models. We compared natural killer (NK) cells from young and aged mice to investigate age-related defects in NK cell distribution and development. Our findings indicate aged mice have reduced NK cells in most peripheral tissues, but not in bone marrow. Reduction of NK cells in periphery was attributed to a reduction of the most mature CD11b(+) CD27(-) NK cells. Apoptosis was not found to explain this specific reduction of mature NK cells. Analysis of NK cell development in bone marrow revealed that aged NK cells progress normally through early stages of development, but a smaller percentage of aged NK cells achieved terminal maturation. Less mature NK cells in aged bone marrow correlated with reduced proliferation of immature NK cells. We propose that advanced age impairs bone marrow maturation of NK cells, possibly affecting homeostasis of NK cells in peripheral tissues. These alterations in NK cell maturational status have critical consequences for NK cell function in advanced age: reduction of the mature circulating NK cells in peripheral tissues of aged mice affects their overall capacity to patrol and eliminate cancerous and viral infected cells.


Subject(s)
Aging , Killer Cells, Natural/cytology , Animals , Apoptosis , Bone Marrow/pathology , Bone Marrow Cells/cytology , CD11b Antigen/metabolism , Cell Proliferation , Cellular Senescence , Male , Mice , Mice, Inbred C57BL , Phenotype , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
20.
Nutr Res ; 33(1): 12-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23351405

ABSTRACT

Administration of bioactive nutritional supplements near or at the time of immunization has been a recent approach to stimulate human immune response to vaccination. Active hexose correlated compound (AHCC), a mushroom extract, has been shown to protect mice against lethal primary influenza infection. Moreover, when AHCC was administered pre-vaccination in mice, they showed improved protection from lethal avian flu infection when compared to mice vaccinated alone. In this study, we hypothesized that AHCC will also improve the immune responses of healthy individuals to influenza vaccine. A randomized controlled study was performed with 30 healthy adults to evaluate the effects of AHCC supplementation on the immune response to the 2009-2010 seasonal influenza vaccine. Blood was drawn pre-vaccination and 3 wk post-vaccination. Immediately post-vaccination, the AHCC group began supplementation with AHCC (3 g/d). Flow cytometric analysis of lymphocyte subpopulations revealed that AHCC supplementation increased NKT cells (P < .1), and CD8 T cells (P < .05) post-vaccination compared to controls. Analysis of antibody production 3 weeks post-vaccination revealed that AHCC supplementation significantly improved protective antibody titers to influenza B, while the improvement was not significant in the control group. Overall, our study showed that AHCC supplementation improved some lymphocyte percentages and influenza B antibody titers over the control. Future studies are required to determine the kinetics of AHCC supplementation to improve the overall response to influenza vaccination.


Subject(s)
Antibody Formation/drug effects , Dietary Supplements , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Polysaccharides/administration & dosage , Agaricales/chemistry , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Humans , Influenza B virus/immunology , Influenza Vaccines/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Middle Aged , Pilot Projects , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL