Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Nat Commun ; 15(1): 1124, 2024 Feb 06.
Article En | MEDLINE | ID: mdl-38321058

The recovery of mitochondrial quality control (MQC) may bring innovative solutions for neuroprotection, while imposing a significant challenge given the need of holistic approaches to restore mitochondrial dynamics (fusion/fission) and turnover (mitophagy and biogenesis). In diabetic retinopathy, this is compounded by our lack of understanding of human retinal neurodegeneration, but also how MQC processes interact during disease progression. Here, we show that mitochondria hyperfusion is characteristic of retinal neurodegeneration in human and murine diabetes, blunting the homeostatic turnover of mitochondria and causing metabolic and neuro-inflammatory stress. By mimicking this mitochondrial remodelling in vitro, we ascertain that N6-furfuryladenosine enhances mitochondrial turnover and bioenergetics by relaxing hyperfusion in a controlled fashion. Oral administration of N6-furfuryladenosine enhances mitochondrial turnover in the diabetic mouse retina (Ins2Akita males), improving clinical correlates and conferring neuroprotection regardless of glycaemic status. Our findings provide translational insights for neuroprotection in the diabetic retina through the holistic recovery of MQC.


Adenosine , Diabetes Mellitus, Experimental , Kinetin , Mitochondrial Dynamics , Male , Mice , Humans , Animals , Neuroprotection , Diabetes Mellitus, Experimental/metabolism , Retina/metabolism , Mitochondria/metabolism
3.
JCI Insight ; 6(12)2021 06 22.
Article En | MEDLINE | ID: mdl-34032637

Evolutionarily conserved signaling intermediate in Toll pathways (ECSIT) is a protein with roles in early development, activation of the transcription factor NF-κB, and production of mitochondrial reactive oxygen species (mROS) that facilitates clearance of intracellular bacteria like Salmonella. ECSIT is also an important assembly factor for mitochondrial complex I. Unlike the murine form of Ecsit (mEcsit), we demonstrate here that human ECSIT (hECSIT) is highly labile. To explore whether the instability of hECSIT affects functions previously ascribed to its murine counterpart, we created a potentially novel transgenic mouse in which the murine Ecsit gene is replaced by the human ECSIT gene. The humanized mouse has low levels of hECSIT protein, in keeping with its intrinsic instability. Whereas low-level expression of hECSIT was capable of fully compensating for mEcsit in its roles in early development and activation of the NF-κB pathway, macrophages from humanized mice showed impaired clearance of Salmonella that was associated with reduced production of mROS. Notably, severe cardiac hypertrophy was manifested in aging humanized mice, leading to premature death. The cellular and molecular basis of this phenotype was delineated by showing that low levels of human ECSIT protein led to a marked reduction in assembly and activity of mitochondrial complex I with impaired oxidative phosphorylation and reduced production of ATP. Cardiac tissue from humanized hECSIT mice also showed reduced mitochondrial fusion and more fission but impaired clearance of fragmented mitochondria. A cardiomyocyte-intrinsic role for Ecsit in mitochondrial function and cardioprotection is also demonstrated. We also show that cardiac fibrosis and damage in humans correlated with low expression of human ECSIT. In summary, our findings identify a role for ECSIT in cardioprotection, while generating a valuable experimental model to study mitochondrial dysfunction and cardiac pathophysiology.


Adaptor Proteins, Signal Transducing , Cardiomegaly , Myocardium , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Humans , Macrophages/metabolism , Mice , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/genetics , NF-kappa B/metabolism
4.
J Clin Endocrinol Metab ; 106(1): 26-41, 2021 01 01.
Article En | MEDLINE | ID: mdl-32617576

CONTEXT: Preeclampsia is a leading cardiovascular complication in pregnancy lacking effective diagnostic and treatment strategies. OBJECTIVE: To investigate the diagnostic and therapeutic target potential of the angiogenesis proteins, FK506-binding protein like (FKBPL) and CD44. DESIGN AND INTERVENTION: FKBPL and CD44 plasma concentration or placental expression were determined in women pre- or postdiagnosis of preeclampsia. Trophoblast and endothelial cell function was assessed following mesenchymal stem cell (MSC) treatment and in the context of FKBPL signaling. SETTINGS AND PARTICIPANTS: Human samples prediagnosis (15 and 20 weeks of gestation; n ≥ 57), or postdiagnosis (n = 18 for plasma; n = 4 for placenta) of preeclampsia were used to determine FKBPL and CD44 levels, compared to healthy controls. Trophoblast or endothelial cells were exposed to low/high oxygen, and treated with MSC-conditioned media (MSC-CM) or a FKBPL overexpression plasmid. MAIN OUTCOME MEASURES: Preeclampsia risk stratification and diagnostic potential of FKBPL and CD44 were investigated. MSC treatment effects and FKBPL-CD44 signaling in trophoblast and endothelial cells were assessed. RESULTS: The CD44/FKBPL ratio was reduced in placenta and plasma following clinical diagnosis of preeclampsia. At 20 weeks of gestation, a high plasma CD44/FKBPL ratio was independently associated with the 2.3-fold increased risk of preeclampsia (odds ratio = 2.3, 95% confidence interval [CI] 1.03-5.23, P = 0.04). In combination with high mean arterial blood pressure (>82.5 mmHg), the risk further increased to 3.9-fold (95% CI 1.30-11.84, P = 0.016). Both hypoxia and MSC-based therapy inhibited FKBPL-CD44 signaling, enhancing cell angiogenesis. CONCLUSIONS: The FKBPL-CD44 pathway appears to have a central role in the pathogenesis of preeclampsia, showing promising utilities for early diagnostic and therapeutic purposes.


Hyaluronan Receptors/physiology , Mesenchymal Stem Cell Transplantation , Pre-Eclampsia , Tacrolimus Binding Proteins/physiology , Adult , Biomarkers/analysis , Case-Control Studies , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/genetics , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Pre-Eclampsia/diagnosis , Pre-Eclampsia/genetics , Pre-Eclampsia/therapy , Pregnancy , Prognosis , Risk Assessment , Signal Transduction/genetics , Tacrolimus Binding Proteins/analysis , Tacrolimus Binding Proteins/genetics , Young Adult
5.
Arterioscler Thromb Vasc Biol ; 40(12): e367-e379, 2020 12.
Article En | MEDLINE | ID: mdl-33115265

OBJECTIVE: Myeloid cells are critically involved in inflammation-induced angiogenesis, although their pathogenic role in the ischemic retina remains controversial. We hypothesize that myeloid cells contribute to pathogenic neovascularization in retinopathy of prematurity through STAT3 (signal transducer and activator of transcription 3) activation. Approach and Results: Using the mouse model of oxygen-induced retinopathy, we show that myeloid cells (CD45+IsolectinB4 [IB4]+) and particularly M2-type macrophages (CD45+ Arg1+), comprise a major source of STAT3 activation (pSTAT3) in the immature ischemic retina. Most of the pSTAT3-expressing myeloid cells concentrated at the hyaloid vasculature and their numbers were strongly correlated with the severity of pathogenic neovascular tuft formation. Pharmacological inhibition of STAT3 reduced the load of IB4+ cells in the hyaloid vasculature and significantly reduced the formation of pathogenic neovascular tufts during oxygen-induced retinopathy, leading to improved long-term visual outcomes (ie, increased retinal thickness and scotopic b-wave electroretinogram responses). Genetic deletion of SOCS3 (suppressor of cytokine signaling 3), an endogenous inhibitor of STAT3, in myeloid cells, enhanced pathological and physiological neovascularization in oxygen-induced retinopathy, indicating that myeloid-STAT3 signaling is crucial for retinal angiogenesis. CONCLUSIONS: Circulating myeloid cells may migrate to the immature ischemic retina through the hyaloid vasculature and contribute to retinal neovascularization via activation of STAT3. Understanding how STAT3 modulates myeloid cells for vascular repair/pathology may provide novel therapeutic options in pathogenic angiogenesis.


Macrophages/metabolism , Oxygen , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , STAT3 Transcription Factor/metabolism , Animals , Animals, Newborn , Anthraquinones/pharmacology , Disease Models, Animal , Female , Macrophages/drug effects , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/prevention & control , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction , Sulfonamides/pharmacology , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 117(30): 18018-18028, 2020 07 28.
Article En | MEDLINE | ID: mdl-32651278

CCN3 is a matricellular protein that promotes oligodendrocyte progenitor cell differentiation and myelination in vitro and ex vivo. CCN3 is therefore a candidate of interest in central nervous system (CNS) myelination and remyelination, and we sought to investigate the expression and role of CCN3 during these processes. We found CCN3 to be expressed predominantly by neurons in distinct areas of the CNS, primarily the cerebral cortex, hippocampus, amygdala, suprachiasmatic nuclei, anterior olfactory nuclei, and spinal cord gray matter. CCN3 was transiently up-regulated following demyelination in the brain of cuprizone-fed mice and spinal cord lesions of mice injected with lysolecithin. However, CCN3-/- mice did not exhibit significantly different numbers of oligodendroglia or differentiated oligodendrocytes in the healthy or remyelinating CNS, compared to WT controls. These results suggest that despite robust and dynamic expression in the CNS, CCN3 is not required for efficient myelination or remyelination in the murine CNS in vivo.


Central Nervous System/metabolism , Demyelinating Diseases/etiology , Gene Expression Regulation , Nephroblastoma Overexpressed Protein/genetics , Remyelination/genetics , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Mice , Myelin Sheath/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
7.
JCI Insight ; 4(23)2019 12 05.
Article En | MEDLINE | ID: mdl-31661466

Mitochondrial quality control (MQC) is crucial for regulating CNS homeostasis, and its disruption has been implicated in the pathogenesis of some of the most common neurodegenerative diseases. In healthy tissues, the maintenance of MQC depends upon an exquisite balance between mitophagy (removal of damaged mitochondria by autophagy) and biogenesis (de novo synthesis of mitochondria). Here, we show that mitophagy is disrupted in diabetic retinopathy (DR) and decoupled from mitochondrial biogenesis during the progression of the disease. Diabetic retinas from human postmortem donors and experimental mice exhibit a net loss of mitochondrial contents during the early stages of the disease process. Using diabetic mitophagy-reporter mice (mitoQC-Ins2Akita) alongside pMitoTimer (a molecular clock to address mitochondrial age dynamics), we demonstrate that mitochondrial loss arose due to an inability of mitochondrial biogenesis to compensate for diabetes-exacerbated mitophagy. However, as diabetes duration increases, Pink1-dependent mitophagy deteriorates, leading to the build-up of mitochondria primed for degradation in DR. Impairment of mitophagy during prolonged diabetes is linked with the development of retinal senescence, a phenotype that blunted hyperglycemia-induced mitophagy in mitoQC primary Müller cells. Our findings suggest that normalizing mitochondrial turnover may preserve MQC and provide therapeutic options for the management of DR-associated complications.


Diabetic Retinopathy/metabolism , Mitochondria/metabolism , Mitophagy/physiology , Animals , Cell Line , Diabetes Mellitus , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Disease Models, Animal , Disease Progression , Female , Humans , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondrial Dynamics/physiology , Mitophagy/genetics , Protein Kinases/metabolism , Retina/metabolism
8.
Diab Vasc Dis Res ; 16(5): 440-449, 2019 09.
Article En | MEDLINE | ID: mdl-31023085

The purpose of this study was to understand the influence of sustained intravitreal vascular endothelial growth factor neutralisation on the retinal and choroidal vasculature in diabetic eyes. Ins2Akita diabetic mice received five intravitreal injections of anti-mouse vascular endothelial growth factor antibody or goat immunoglobulin G (0.2 µg/µL/eye) over a 4-month period. Retinal and choroidal vascular changes were analysed by confocal microscopy of tissue flat-mounts. Retinal gene expression of vascular endothelial growth factor family members (vascular endothelial growth factors A, B, C and D), vascular endothelial growth factor receptors (sVEGFR-1 and VEGFR-2) and tight junctions (claudin 1, 2, 5; occludin and zonula occludens-1) were analysed by quantitative reverse transcription polymerase chain reaction. Vascular endothelial growth factor A and claudin 5 were significantly increased in diabetic retinae. Gene expression was unaffected by anti-vascular endothelial growth factor treatment. The number of acellular vessels was increased in diabetic retinae and reduced following anti-vascular endothelial growth factor treatment. Retinal and choroidal vascular density and area were unaffected by sustained vascular endothelial growth factor neutralisation. Our results suggest that five consecutive intravitreal anti-vascular endothelial growth factor injections do not cause significant vascular changes in the retina and choroid in diabetic and non-diabetic mice.


Angiogenesis Inhibitors/administration & dosage , Antibodies, Neutralizing/administration & dosage , Choroid/blood supply , Choroidal Neovascularization , Diabetes Mellitus/drug therapy , Diabetic Retinopathy/prevention & control , Retinal Neovascularization , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/toxicity , Animals , Antibodies, Neutralizing/toxicity , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Gene Expression Regulation , Intravitreal Injections , Male , Mice, Inbred C57BL , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Diabetologia ; 61(11): 2433-2446, 2018 11.
Article En | MEDLINE | ID: mdl-30019207

AIMS/HYPOTHESIS: Cataract surgery in diabetic individuals worsens pre-existing retinopathy and triggers the development of diabetic ocular complications, although the underlying cellular and molecular pathophysiology remains elusive. We hypothesise that lens surgery may exaggerate pre-existing retinal inflammation in diabetes, which may accelerate neurovascular degeneration in diabetic eyes. METHODS: Male heterozygous Ins2Akita mice (3 months of age) and C57BL/6 J age-matched siblings received either lens capsulotomy (to mimic human cataract surgery) or corneal incision (sham surgery) in the right eye. At different days post surgery, inflammation in anterior/posterior ocular tissues was assessed by immunohistochemistry and proinflammatory gene expression in the retina by quantitative PCR (qPCR). Degenerative changes in the retina were evaluated by electroretinography, in vivo examination of retinal thickness (using spectral domain optical coherence tomography [SD-OCT]) and morphometric analysis of retinal neurons. The therapeutic benefit of neutralising Wnt/ß-catenin signalling following lens capsulotomy was evaluated by intravitreal administration of monoclonal antibody against the co-receptor low-density lipoprotein receptor-related protein 6 (LRP6) (Mab2F1; 5 µg/µl in each eye). RESULTS: Lens capsulotomy triggered the early onset of retinal neurodegeneration in Ins2Akita mice, evidenced by abnormal scotopic a- and b-wave responses, reduced retinal thickness and degeneration of outer/inner retinal neurons. Diabetic Ins2Akita mice also had a higher number of infiltrating ionised calcium-binding adapter molecule 1 (IBA1)/CD68+ cells in the anterior/posterior ocular tissues and increased retinal expression of inflammatory mediators (chemokine [C-C motif] ligand 2 [CCL2] and IL-1ß). The expression of ß-catenin was significantly increased in the inner nuclear layer, ganglion cells and infiltrating immune cells in Ins2Akita mice receiving capsulotomy. Neutralisation of Wnt/ß-catenin signalling by Mab2F1 ameliorated ocular inflammation and prevented capsulotomy-induced retinal degeneration in the Ins2Akita mouse model of diabetes. CONCLUSIONS/INTERPRETATION: Targeting the canonical Wnt/ß-catenin signalling pathway may provide a novel approach for the postoperative management of diabetic individuals needing cataract surgery.


Cataract Extraction/adverse effects , Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/pathology , Disease Models, Animal , Electroretinography , Immunoblotting , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Retinal Degeneration/pathology
10.
Am J Physiol Renal Physiol ; 312(6): F1141-F1157, 2017 06 01.
Article En | MEDLINE | ID: mdl-28100499

Gremlin1 (Grem1), an antagonist of bone morphogenetic proteins, plays a key role in embryogenesis. A highly specific temporospatial gradient of Grem1 and bone morphogenetic protein signaling is critical to normal lung, kidney, and limb development. Grem1 levels are increased in renal fibrotic conditions, including acute kidney injury, diabetic nephropathy, chronic allograft nephropathy, and immune glomerulonephritis. We demonstrate that a small number of grem1-/- whole body knockout mice on a mixed genetic background (8%) are viable, with a single, enlarged left kidney and grossly normal histology. The grem1-/- mice displayed mild renal dysfunction at 4 wk, which recovered by 16 wk. Tubular epithelial cell-specific targeted deletion of Grem1 (TEC-grem1-cKO) mice displayed a milder response in the acute injury and recovery phases of the folic acid model. Increases in indexes of kidney damage were smaller in TEC-grem1-cKO than wild-type mice. In the recovery phase of the folic acid model, associated with renal fibrosis, TEC-grem1-cKO mice displayed reduced histological damage and an attenuated fibrotic gene response compared with wild-type controls. Together, these data demonstrate that Grem1 expression in the tubular epithelial compartment plays a significant role in the fibrotic response to renal injury in vivo.


Acute Kidney Injury/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Tubules/metabolism , Urogenital Abnormalities/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Disease Models, Animal , Female , Fibrosis , Folic Acid , Gene Expression Regulation , Genetic Predisposition to Disease , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules/abnormalities , Kidney Tubules/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Organogenesis , Phenotype , Signal Transduction , Time Factors , Urogenital Abnormalities/genetics , Urogenital Abnormalities/pathology
11.
Sci Rep ; 5: 18316, 2015 Dec 16.
Article En | MEDLINE | ID: mdl-26671074

Current therapies that target vascular endothelial growth factor (VEGF) have become a mainstream therapy for the management of diabetic macular oedema. The treatment involves monthly repeated intravitreal injections of VEGF inhibitors. VEGF is an important growth factor for many retinal cells, including different types of neurons. In this study, we investigated the adverse effect of multiple intravitreal anti-VEGF injections (200 ng/µl/eye anti-mouse VEGF164, once every 2 weeks totalling 5-6 injections) to retinal neurons in Ins2(Akita) diabetic mice. Funduscopic examination revealed the development of cotton wool spot-like lesions in anti-VEGF treated Ins2(Akita) mice after 5 injections. Histological investigation showed focal swellings of retinal nerve fibres with neurofilament disruption. Furthermore, anti-VEGF-treated Ins2(Akita) mice exhibited impaired electroretinographic responses, characterized by reduced scotopic a- and b-wave and oscillatory potentials. Immunofluorescent staining revealed impairment of photoreceptors, disruptions of synaptic structures and loss of amacrine and retinal ganglion cells in anti-VEGF treated Ins2(Akita) mice. Anti-VEGF-treated WT mice also presented mild amacrine and ganglion cell death, but no overt abnormalities in photoreceptors and synaptic structures. At the vascular level, exacerbated albumin leakage was observed in anti-VEGF injected diabetic mice. Our results suggest that sustained intraocular VEGF neutralization induces retinal neurodegeneration and vascular damage in the diabetic eye.


Antibodies, Neutralizing/adverse effects , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Insulin/deficiency , Neurodegenerative Diseases/chemically induced , Retinal Neurons/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Neutralizing/pharmacology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Mice , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Retinal Neurons/pathology , Vascular Endothelial Growth Factor A/metabolism
12.
PLoS One ; 9(5): e97970, 2014.
Article En | MEDLINE | ID: mdl-24848689

Retinal neurodegeneration is a key component of diabetic retinopathy (DR), although the detailed neuronal damage remains ill-defined. Recent evidence suggests that in addition to amacrine and ganglion cell, diabetes may also impact on other retinal neurons. In this study, we examined retinal degenerative changes in Ins2Akita diabetic mice. In scotopic electroretinograms (ERG), b-wave and oscillatory potentials were severely impaired in 9-month old Ins2Akita mice. Despite no obvious pathology in fundoscopic examination, optical coherence tomography (OCT) revealed a progressive thinning of the retina from 3 months onwards. Cone but not rod photoreceptor loss was observed in 3-month-old diabetic mice. Severe impairment of synaptic connectivity at the outer plexiform layer (OPL) was detected in 9-month old Ins2Akita mice. Specifically, photoreceptor presynaptic ribbons were reduced by 25% and postsynaptic boutons by 70%, although the density of horizontal, rod- and cone-bipolar cells remained similar to non-diabetic controls. Significant reductions in GABAergic and glycinergic amacrine cells and Brn3a+ retinal ganglion cells were also observed in 9-month old Ins2Akita mice. In conclusion, the Ins2Akita mouse develops cone photoreceptor degeneration and the impairment of synaptic connectivity at the OPL, predominately resulting from the loss of postsynaptic terminal boutons. Our findings suggest that the Ins2Akita mouse is a good model to study diabetic retinal neuropathy.


Diabetic Retinopathy/pathology , Retinal Neurons/pathology , Synapses/pathology , Animals , Diabetes Mellitus, Type 1/complications , Diabetic Retinopathy/complications , Electroretinography , Male , Mice , Time Factors
13.
Exp Eye Res ; 124: 1-10, 2014 Jul.
Article En | MEDLINE | ID: mdl-24792588

Insulin receptor substrate-2 (Irs2) mediates peripheral insulin action and is essential for retinal health. Previous investigations have reported severe photoreceptor degeneration and abnormal visual function in Irs2-deficient mice. However, molecular changes in the Irs2(-)(/)(-) mouse retina have not been described. In this study, we examined retinal degenerative changes in neuronal and glial cells of adult (9- and 12-week old) Irs2(-)(/)(-) mice by immunohistochemistry. 9-week old Irs2(-)(/)(-) mice showed significant thinning of outer retinal layers, concomitant to Müller and microglial cell activation. Photoreceptor cells displayed different signs of degeneration, such as outer/inner segment atrophy, redistribution of rod- and cone-opsins and spatial disorganization of cone cells. This was accompanied by synaptic changes at the outer plexiform layer, including the retraction of rod-spherules, reduction of photoreceptor synaptic ribbons and synaptic remodeling in second order neurons (i.e. loss and sprouting of dendritic processes in rod bipolar and horizontal cells). By 12 weeks of age, the thickness of inner retinal layers was severely affected. Although inner plexiform layer stratification remained unchanged at this stage, rod bipolar cell axon terminals were significantly depleted. Significant loss of Brn3a(+) retinal ganglion cells occurred in 12-week old Irs2(-)(/)(-) mice, in contrast to younger ages. Adult Irs2(-)(/)(-) mice showed clear hallmarks of neurodegeneration and disruption of the inner retina with increasing age. Pharmacological stimulation of Irs2 signaling pathway may provide additional neuroprotection in certain degenerative retinopathies.


Insulin Receptor Substrate Proteins/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Bipolar Cells/pathology , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology , Vision, Ocular/physiology , Animals , Disease Models, Animal , Electroretinography , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Photoreceptor Cells, Vertebrate/metabolism , Retinal Bipolar Cells/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/metabolism
14.
Transl Vis Sci Technol ; 3(2): 3, 2014 Mar.
Article En | MEDLINE | ID: mdl-24757593

PURPOSE: To investigate the adverse effect of intravitreal injection of normal saline (NS) and phosphate buffered saline (PBS) in mouse eyes. METHODS: NS or PBS was injected intravitreally into C57BL/6J mouse eyes. Retinal lesions were monitored by fundus imaging, spectral-domain optical coherence tomography (SD-OCT), and histological investigations. Retinal immune gene expression was determined by real-time polymerase chain reaction (PCR). The toxic effect of NS and PBS or retinal protein from NS- or PBS-injected eyes on retinal pigment epithelium (RPE) was tested in B6-RPE-07 mouse RPE cell cultures. RESULTS: Intravitreal injection of NS dose-dependently induced localized retinal lesion in mice. Histological investigations revealed multiple vacuoles in photoreceptor outer segments and RPE cells. The lesions recovered over time and by 3 weeks post injection the majority of lesions vanished in eyes receiving 1 µl NS. Inflammatory genes, including TNF-α, IL-1ß, IL-6, iNOS, and VEGF were upregulated in NS injected eyes. Intravitreal injection of PBS did not cause any pathology. The treatment of B6-RPE07 cells with 30% PBS or 30% NS did not affect RPE viability. However, incubation of 1-µg/ml retinal protein from NS-injected eyes, but not PBS-injected eyes induced RPE cell death. CONCLUSION: NS is toxic to the C57BL/6J mouse retina and should not be used as a vehicle for intraocular injection. PBS is not toxic to the retina and is a preferred vehicle. TRANSLATIONAL RELEVANCE: NS is not a physiological solution for intraocular injection in the C57BL/6J mice and questions its suitability for intraocular injection in other species, including human.

15.
PLoS One ; 8(4): e61381, 2013.
Article En | MEDLINE | ID: mdl-23637822

Previous studies have shown that CCL2/CX3CR1 deficient mice on C57BL/6N background (with rd8 mutation) have an early onset (6 weeks) of spontaneous retinal degeneration. In this study, we generated CCL2(-/-)CX3CR1(GFP/GFP) mice on the C57BL/6J background. Retinal degeneration was not detected in CCL2(-/-)CX3CR1(GFP/GFP) mice younger than 6 months. Patches of whitish/yellowish fundus lesions were observed in 17∼60% of 12-month, and 30∼100% of 18-month CCL2(-/-)CX3CR1(GFP/GFP) mice. Fluorescein angiography revealed no choroidal neovascularisation in these mice. Patches of retinal pigment epithelium (RPE) and photoreceptor damage were detected in 30% and 50% of 12- and 18-month CCL2(-/-)CX3CR1(GFP/GFP) mice respectively, but not in wild-type mice. All CCL2(-/-)CX3CR1(GFP/GFP) mice exposed to extra-light (∼800lux, 6 h/day, 6 months) developed patches of retinal atrophy, and only 20-25% of WT mice which underwent the same light treatment developed atrophic lesions. In addition, synaptophysin expression was detected in the outer nucler layer (ONL) of area related to photoreceptor loss in CCL2(-/-)CX3CR1(GFP/GFP) mice. Markedly increased rhodopsin but reduced cone arrestin expression was observed in retinal outer layers in aged CCL2(-/-)CX3CR1(GFP/GFP) mice. GABA expression was reduced in the inner retina of aged CCL2(-/-)CX3CR1(GFP/GFP) mice. Significantly increased Müller glial and microglial activation was observed in CCL2(-/-)CX3CR1(GFP/GFP) mice compared to age-matched WT mice. Macrophages from CCL2(-/-)CX3CR1(GFP/GFP) mice were less phagocytic, but expressed higher levels of iNOS, IL-1ß, IL-12 and TNF-α under hypoxia conditions. Our results suggest that the deletions of CCL2 and CX3CR1 predispose mice to age- and light-mediated retinal damage. The CCL2/CX3CR1 deficient mouse may thus serve as a model for age-related atrophic degeneration of the RPE, including the dry type of macular degeneration, geographic atrophy.


Chemokine CCL2/deficiency , Macular Degeneration/pathology , Receptors, Cytokine/deficiency , Receptors, HIV/deficiency , Retina/physiology , Retinal Degeneration/pathology , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/genetics , Green Fluorescent Proteins/metabolism , Light , Macrophages/physiology , Macular Degeneration/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/physiology , Receptors, Cytokine/genetics , Receptors, HIV/genetics , Retina/pathology
16.
Cell Tissue Res ; 350(1): 183-7, 2012 Oct.
Article En | MEDLINE | ID: mdl-22777743

Glutamine synthetase (GS) catalyzes the de novo synthesis of glutamine, an amino acid that has been shown to influence sperm motility in mammals. To date, no information is available about GS content in human sperm. In this study, we have characterized the presence and cellular location of GS in fresh human normozoospermic samples. We have detected a single band corresponding to GS by Western blot. Confocal analysis has revealed GS immunoreactivity in the post-acrosomal head region. Moreover, double-labeling experiments with either F-actin or calicin have demonstrated GS confinement in the post-acrosomal region of the perinuclear theca. These data have been validated by a post-embedding ultra-structural study. The presence of GS in the post-acrosomal region of the perinuclear theca suggests that human sperm can carry out in glutamine synthesis.


Glutamate-Ammonia Ligase/metabolism , Spermatozoa/enzymology , Animals , Blotting, Western , Glutamate-Ammonia Ligase/ultrastructure , Humans , Male , Mice , Mice, Inbred C57BL , Protein Transport , Spermatozoa/cytology , Spermatozoa/ultrastructure , Tissue Extracts
17.
Brain Res ; 1432: 46-55, 2012 Jan 13.
Article En | MEDLINE | ID: mdl-22138430

Aquaporins (AQPs) are membrane proteins that facilitate water transport across biological membranes and are essential for the proper function of neural tissue. Although AQPs have been extensively studied in mammalian retina, their presence in lower vertebrate retina is less frequently characterized. AQP4 expressed in mammalian and chick Müller cells plays a major part in maintaining retinal homeostasis. In this study, we examined the immunoreactivity of AQP4 in the adult retina of gilthead sea bream (Sparus aurata-teleost fish), during light and dark adaptation. The AQP4 expression was detected in Müller cell somas at the inner nuclear layer and in the end-feet processes near the vitreoretinal border. Moreover, AQP4 was also evident in cone photoreceptor cells and in a GABAergic subpopulation of amacrine cells (AQP4-ACs). Four different types of AQP4-ACs were characterized based on their morphology and dendrite stratification. Interestingly, a stronger AQP4 immunoreactivity was observed in the inner nuclear layer during dark adaptation, accompanied by a significant increment in AQP4-ACs cell size. Hence, AQP4 may play an important role in water distribution in the teleost fish retina.


Amacrine Cells/metabolism , Aquaporin 4/metabolism , Fishes/immunology , Retina/metabolism , Adaptation, Ocular/physiology , Amacrine Cells/cytology , Amacrine Cells/immunology , Animals , Aquaporin 4/physiology , Body Water/metabolism , Cell Shape/physiology , Dark Adaptation/physiology , Dendrites/metabolism , Dendrites/ultrastructure , Ecosystem , Neuroglia/cytology , Neuroglia/immunology , Neuroglia/metabolism , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Species Specificity , Vision, Ocular/physiology
18.
Brain Res ; 1321: 60-6, 2010 Mar 19.
Article En | MEDLINE | ID: mdl-20114037

The S334ter-3 rat is a transgenic model of retinal degeneration (RD) developed to express a rhodopsin mutation similar to that found in human retinitis pigmentosa. Due to this advantage over other models of RD, a few retina transplant studies have been reported on this animal model. Currently, no information is available on cone photoreceptor changes that occur in the S334ter RD model. In this study, we investigated the effect of RD on the morphology, distribution, and synaptic connectivity of short-wavelength cones (S-cones) during development of S334ter-3 rat retinas. At P21 RD retinas, the outer-nuclear layer was significantly narrower, while S-cones showed shortening of their segments and axons compared to control retinas. From P90 onward, S-opsin-immunoreactive cells appeared at the outer margin of the inner-nuclear layer of RD retinas. Double-labelling experiments showed these cells contained recoverin and cone arrestin. Furthermore, ultra-structure study showed that synaptic ribbons are conserved in the S-cone at P180 RD retinas. Although cell density of S-cones significantly dropped after P90, survival rates depended on the retinal region. Overall, the S334ter-3 RD model shows hallmarks of cone remodelling due to photoreceptor degeneration.


Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/pathology , Animals , Immunohistochemistry , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism
...