Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(4): 522-536, 2023 04.
Article in English | MEDLINE | ID: mdl-36794587

ABSTRACT

BACKGROUND: CD4+ (cluster of differentation) and CD8+ T cells are increased in the ocular fluids of patients with neovascular retinopathy, yet their role in the disease process is unknown. METHODS: We describe how CD8+ T cells migrate into the retina and contribute to pathological angiogenesis by releasing cytokines and cytotoxic factors. RESULTS: In oxygen-induced retinopathy, flow cytometry revealed the numbers of CD4+ and CD8+ T cells were increased in blood, lymphoid organs, and retina throughout the development of neovascular retinopathy. Interestingly, the depletion of CD8+ T cells but not CD4+ T cells reduced retinal neovascularization and vascular leakage. Using reporter mice expressing gfp (green fluorescence protein) in CD8+ T cells, these cells were localized near neovascular tufts in the retina, confirming that CD8+ T cells contribute to the disease. Furthermore, the adoptive transfer of CD8+ T cells deficient in TNF (tumor necrosis factor), IFNγ (interferon gamma), Prf (perforin), or GzmA/B (granzymes A/B) into immunocompetent Rag1-/- mice revealed that CD8+ T cells mediate retinal vascular disease via these factors, with TNF influencing all aspects of vascular pathology. The pathway by which CD8+ T cells migrate into the retina was identified as CXCR3 (C-X-C motif chemokine receptor 3) with the CXCR3 blockade reducing the number of CD8+ T cells within the retina and retinal vascular disease. CONCLUSIONS: We discovered that CXCR3 is central to the migration of CD8+ T cells into the retina as the CXCR3 blockade reduced the number of CD8+ T cells within the retina and vasculopathy. This research identified an unappreciated role for CD8+ T cells in retinal inflammation and vascular disease. Reducing CD8+ T cells via their inflammatory and recruitment pathways is a potential treatment for neovascular retinopathies.


Subject(s)
Retinal Diseases , Vascular Diseases , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , Neovascularization, Pathologic , Retina/metabolism , Retinal Diseases/metabolism , Interferon-gamma/metabolism , Vascular Diseases/pathology , Mice, Inbred C57BL
2.
Int J Mol Sci ; 24(3)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36768656

ABSTRACT

Vision loss in diabetic retinopathy features damage to the blood-retinal barrier and neovascularization, with hypertension and the renin-angiotensin system (RAS) having causal roles. We evaluated if finerenone, a non-steroidal mineralocorticoid receptor (MR) antagonist, reduced vascular pathology and inflammation in diabetic and neovascular retinopathy. Diabetic and hypertensive transgenic (mRen-2)27 rats overexpressing the RAS received the MR antagonist finerenone (10 mg/kg/day, oral gavage) or the angiotensin-converting enzyme inhibitor perindopril (10 mg/kg/day, drinking water) for 12 weeks. As retinal neovascularization does not develop in diabetic rodents, finerenone (5 mg/kg/day, i.p.) was evaluated in murine oxygen-induced retinopathy (OIR). Retinal vasculopathy was assessed by measuring gliosis, vascular leakage, neovascularization, and VEGF. Inflammation was investigated by quantitating retinal microglia/macrophages, pro-inflammatory mediators, and anti-inflammatory regulatory T-cells (Tregs). In diabetes, both treatments reduced systolic blood pressure, gliosis, vascular leakage, and microglial/macrophage density, but only finerenone lowered VEGF, ICAM-1, and IL-1ß. In OIR, finerenone reduced neovascularization, vascular leakage, and microglial density, and increased Tregs in the blood, spleen, and retina. Our findings, in the context of the FIDELIO-DKD and FIGARO-DKD trials reporting the benefits of finerenone on renal and cardiovascular outcomes in diabetic kidney disease, indicate the potential of finerenone as an effective oral treatment for diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Diabetic Retinopathy , Vascular System Injuries , Rats , Animals , Mice , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/complications , Rodentia , Gliosis/complications , Vascular Endothelial Growth Factor A , T-Lymphocytes, Regulatory , Naphthyridines/pharmacology , Diabetic Nephropathies/etiology , Neovascularization, Pathologic/complications , Inflammation/complications , Diabetes Mellitus, Type 2/complications
3.
Int J Mol Sci ; 24(21)2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37958664

ABSTRACT

Retinal inflammation is a central feature of ocular neovascular diseases such as diabetic retinopathy and retinopathy of prematurity, but the contribution of neutrophils to this process is not fully understood. We studied oxygen-induced retinopathy (OIR) which develops in two phases, featuring hyperoxia-induced retinal vaso-obliteration in phase I, followed by retinal neovascularization in phase II. As neutrophils are acute responders to tissue damage, we evaluated whether neutrophil depletion with an anti-Ly6G mAb administered in phase I OIR influenced retinal inflammation and vascular injury. Neutrophils were measured in blood and spleen via flow cytometry, and myeloperoxidase, an indicator of neutrophil activity, was evaluated in the retina using Western blotting. Retinal vasculopathy was assessed by quantitating vaso-obliteration, neovascularization, vascular leakage, and VEGF levels. The inflammatory factors, TNF, MCP-1, and ICAM-1 were measured in retina. In the OIR controls, neutrophils were increased in the blood and spleen in phase I but not phase II OIR. In OIR, the anti-Ly6G mAb reduced neutrophils in the blood and spleen, and myeloperoxidase, inflammation, and vasculopathy in the retina. Our findings revealed that the early rise in neutrophils in OIR primes the retina for an inflammatory and angiogenic response that promotes severe damage to the retinal vasculature.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Mice , Oxygen/adverse effects , Neutrophils , Peroxidase , Retinopathy of Prematurity/chemically induced , Vascular Endothelial Growth Factor A/physiology , Animals, Newborn , Retina , Neovascularization, Pathologic , Inflammation , Mice, Inbred C57BL , Disease Models, Animal
4.
Immunity ; 39(3): 573-83, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24012421

ABSTRACT

Activation-induced cell death (AICD) plays a critical role in immune homeostasis and tolerance. In T-cell-dependent humoral responses, AICD of B cells is initiated by Fas ligand (FasL) on T cells, stimulating the Fas receptor on B cells. In contrast, T-cell-independent B cell responses involve innate-type B lymphocytes, such as marginal zone (MZ) B cells, and little is known about the mechanisms that control AICD during innate B cell responses to Toll-like receptor (TLR) activation. Here, we show that MZ B cells undergo AICD in response to TLR4 activation in vivo. The transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor and TLR4 cooperate to upregulate expression of both FasL and Fas on MZ B cells and also to repress inhibitors of Fas-induced apoptosis signaling. These findings demonstrate an unappreciated role for TACI and its ligands in the regulation of AICD during T-cell-independent B cell responses.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Toll-Like Receptor 4/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , fas Receptor/metabolism , Animals , B-Cell Activation Factor Receptor/biosynthesis , B-Lymphocytes/immunology , Enzyme Activation , Fas Ligand Protein/biosynthesis , Lipopolysaccharides , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Transmembrane Activator and CAML Interactor Protein/genetics
5.
Am J Pathol ; 190(9): 1801-1812, 2020 09.
Article in English | MEDLINE | ID: mdl-32526165

ABSTRACT

Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are two debilitating disorders that develop in preterm infants exposed to supplemental oxygen to prevent respiratory failure. Both can lead to lifelong disabilities, such as chronic obstructive pulmonary disease and vision loss. Due to the lack of a standard experimental model of coincident disease, the underlying associations between BPD and ROP are not well characterized. To address this gap, we used the robust mouse model of oxygen-induced retinopathy exposing C57BL/6 mice to 75% oxygen from postnatal day 7 to 12. The cardinal features of ROP were replicated by this strategy, and the lungs of the same mice were simultaneously examined for evidence of BPD-like lung injury, investigating both the short- and long-term effects of early-life supplemental oxygen exposure. At postnatal days 12 and 18, mild lung disease was evident by histopathologic analysis together with the expected vasculopathy in the inner retina. At later time points, the lung lesion had progressed to severe airspace enlargement and alveolar simplification, with concurrent thinning in the outer layer of the retina. In addition, critical angiogenic oxidative stress and inflammatory factors reported to be dysregulated in ROP were similarly impaired in the lungs. These data shed new light on the interconnectedness of these two neonatal disorders, holding potential for the discovery of novel targets to treat BPD and ROP.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Disease Models, Animal , Oxygen Inhalation Therapy/adverse effects , Oxygen/toxicity , Retinopathy of Prematurity/etiology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/pathology , Inflammation/etiology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Retinopathy of Prematurity/pathology
6.
J Neurosci ; 39(28): 5562-5580, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31061088

ABSTRACT

We previously identified that ngr1 allele deletion limits the severity of experimental autoimmune encephalomyelitis (EAE) by preserving axonal integrity. However, whether this favorable outcome observed in EAE is a consequence of an abrogated neuronal-specific pathophysiological mechanism, is yet to be defined. Here we show that, Cre-loxP-mediated neuron-specific deletion of ngr1 preserved axonal integrity, whereas its re-expression in ngr1-/- female mice potentiated EAE-axonopathy. As a corollary, myelin integrity was preserved under Cre deletion in ngr1flx/flx , retinal ganglion cell axons whereas, significant demyelination occurred in the ngr1-/- optic nerves following the re-introduction of NgR1. Moreover, Cre-loxP-mediated axon-specific deletion of ngr1 in ngr1flx/flx mice also demonstrated efficient anterograde transport of fluorescently-labeled ChTxß in the optic nerves of EAE-induced mice. However, the anterograde transport of ChTxß displayed accumulation in optic nerve degenerative axons of EAE-induced ngr1-/- mice, when NgR1 was reintroduced but was shown to be transported efficiently in the contralateral non- recombinant adeno-associated virus serotype 2-transduced optic nerves of these mutant mice. We further identified that the interaction between the axonal motor protein, Kinesin-1 and collapsin response mediator protein 2 (CRMP2) was unchanged upon Cre deletion of ngr1 Whereas, this Kinesin-1/CRMP2 association was reduced when NgR1 was re-expressed in the ngr1-/- optic nerves. Our data suggest that NgR1 governs axonal degeneration in the context of inflammatory-mediated demyelination through the phosphorylation of CRMP2 by stalling axonal vesicular transport. Moreover, axon-specific deletion of ngr1 preserves axonal transport mechanisms, blunting the induction of inflammatory demyelination and limiting the severity of EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is commonly induced by aberrant immune-mediated destruction of the protective sheath of nerve fibers (known as myelin). However, it has been shown that MS lesions do not only consist of this disease pattern, exhibiting heterogeneity with continual destruction of axons. Here we investigate how neuronal NgR1 can drive inflammatory-mediated axonal degeneration and demyelination within the optic nerve by analyzing its downstream signaling events that govern axonal vesicular transport. We identify that abrogating the NgR1/pCRMP2 signaling cascade can maintain Kinesin-1-dependent anterograde axonal transport to limit inflammatory-mediated axonopathy and demyelination. The ability to differentiate between primary and secondary mechanisms of axonal degeneration may uncover therapeutic strategies to limit axonal damage and progressive MS.


Subject(s)
Axonal Transport , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myelin Sheath/metabolism , Nogo Receptor 1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Axons/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Kinesins/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/metabolism , Nogo Receptor 1/genetics , Retinal Ganglion Cells/metabolism , Signal Transduction
7.
Exp Eye Res ; 191: 107902, 2020 02.
Article in English | MEDLINE | ID: mdl-31884019

ABSTRACT

Microglial cells are important contributors to the neuroinflammation and blood vessel damage that occurs in ischemic retinopathies. We hypothesized that key effectors of the renin-angiotensin aldosterone system, angiotensin II (Ang II) and aldosterone, increase the density of microglia in the retina and stimulate their production of reactive oxygen species (ROS) as well as pro-angiogenic and pro-inflammatory factors. Two animal models were studied that featured up-regulation of Ang II or aldosterone and included transgenic Ren-2 rats which overexpress renin and Ang II in tissues including the retina, and Sprague Dawley rats with ischemic retinopathy and infused with aldosterone. Complementary studies were performed in primary cultures of retinal microglia from neonatal Sprague Dawley rats exposed to hypoxia (0.5% O2) and inhibitors of the angiotensin type 1 receptor (valsartan), the mineralocorticoid receptor (spironolactone) or aldosterone synthase (FAD286). In both in vivo models, the density of ionized calcium-binding adaptor protein-1 labelled microglia/macrophages was increased in retina compared to genetic or vehicle controls. In primary cultures of retinal microglia, hypoxia increased ROS (superoxide) levels as well as the expression of the NADPH oxidase (NOX) isoforms, NOX1, NOX2 and NOX4. The elevated levels of ROS as well as NOX2 and NOX4 were reduced by all of the treatments, and valsartan and FAD286 also reduced NOX1 mRNA levels. A protein cytokine array of retinal microglia revealed that valsartan, spironolactone and FAD286 reduced the hypoxia-induced increase in the potent pro-angiogenic and pro-inflammatory agent, vascular endothelial growth factor as well as the inflammatory factors, CCL5 and interferon γ. Valsartan also reduced the hypoxia-induced increase in IL-6 and TIMP-1 as well as the chemoattractants, CXCL2, CXCL3, CXCL5 and CXCL10. Spironolactone and FAD286 reduced the levels of CXCL2 and CXCL10, respectively. In conclusion, our findings that both Ang II and aldosterone influence the activation of retinal microglia implicates the renin-angiotensin aldosterone system in the pathogenesis of ischemic retinopathies.


Subject(s)
Aldosterone/pharmacology , Angiotensin II/pharmacology , Microglia/drug effects , Retinal Neurons/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cytochrome P-450 CYP11B2/metabolism , Female , Immunohistochemistry , Microglia/metabolism , Oxygen/toxicity , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/metabolism , Receptors, Mineralocorticoid/metabolism , Retinal Neovascularization/etiology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neurons/metabolism , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology
8.
Exp Eye Res ; 201: 108255, 2020 12.
Article in English | MEDLINE | ID: mdl-32971094

ABSTRACT

Reactive oxygen species (ROS) normally play an important physiological role in health regulating cellular processes and signal transduction. The amount of ROS is usually kept in fine balance with the generation of ROS largely being offset by the body's antioxidants. A tipping of this balance has increasingly been recognised as a contributor to human disease. The retina, as a result of its cellular anatomy and physical location, is a potent generator of ROS that has been linked to several major retinal diseases. This review will provide a summary of the role of oxidative stress in the pathogenesis of diabetic retinopathy, age-related macular degeneration, myopia, retinal vein occlusion, retinitis pigmentosa and retinopathy of prematurity. Therapies aimed at controlling oxidative stress in these diseases are also examined.


Subject(s)
Antioxidants/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Retina/pathology , Retinal Diseases/metabolism , Animals , Humans , Retina/metabolism , Retinal Diseases/pathology , Signal Transduction
9.
Am J Pathol ; 188(3): 805-817, 2018 03.
Article in English | MEDLINE | ID: mdl-29248456

ABSTRACT

Although increasing evidence indicates that endothelin-2 (Edn2) has distinct roles in tissue pathology, including inflammation, glial cell dysfunction, and angiogenesis, its role in the retina and the factors that regulate its actions are not fully understood. We hypothesized that Edn2 damages the blood-retinal barrier (BRB) and that this is mediated by interactions with the renin-angiotensin-aldosterone system and reactive oxygen species derived from NADPH oxidase (Nox). C57BL/6J mice received an intravitreal injection of Edn2 or control vehicle to examine the blood pressure-independent effects of Edn2. Mice administered Edn2 were randomized to receive by intraperitoneal injection treatments that inhibited the Edn type a receptor, Edn type b receptor, angiotensin type 1 receptor, mineralocorticoid receptor, or Nox isoforms 1 to 4. One month later, mice administered Edn2 exhibited breakdown of the BRB with increased vascular leakage, vascular endothelial growth factor expression, and infiltrating macrophages (Ly6C+CD45highCD11b+). Further, macroglial Müller cells, which influence the integrity of the BRB and prevent retinal edema, became gliotic and expressed increased levels of water (aquaporin-4) and ion (Kir4.1) channels. This Edn2-mediated retinopathy was reduced by all treatments. Complementary in vitro studies in cultured Müller cells supported these findings and demonstrated the importance of reactive oxygen species in mediating these events. In conclusion, Edn2 has detrimental effects on the BRB and Müller cells that involve interactions with the renin-angiotensin aldosterone system and Nox1/4.


Subject(s)
Aldosterone/pharmacology , Angiotensin II/pharmacology , Blood-Retinal Barrier/drug effects , Endothelin-2/pharmacology , Ependymoglial Cells/drug effects , NADPH Oxidases/metabolism , Retina/drug effects , Aquaporin 4/metabolism , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Cell Movement/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Reactive Oxygen Species/metabolism , Retina/metabolism , Retina/pathology
10.
Exp Eye Res ; 187: 107766, 2019 10.
Article in English | MEDLINE | ID: mdl-31425690

ABSTRACT

Angiotensin II and aldosterone are the main effectors of the renin-angiotensin aldosterone system (RAAS) and have a central role in hypertension as well as cardiovascular and renal disease. The localization of RAAS components within the retina has led to studies investigating the roles of angiotensin II, aldosterone and the counter regulatory arm of the pathway in vision-threatening retinopathies. This review will provide a brief overview of RAAS components as well as the vascular pathology that develops in the retinal diseases, retinopathy of prematurity, diabetic retinopathy and neovascular age-related macular degeneration. The review will discuss pre-clinical and clinical evidence that modulation of the RAAS alters the development of vasculopathy and inflammation in the aforementioned retinopathies, as well as the emerging role of aldosterone and the mineralocorticoid receptor in central serous chorioretinopathy.


Subject(s)
Aldosterone/physiology , Angiotensin II/physiology , Diabetic Retinopathy/physiopathology , Retinal Vessels/physiology , Retinitis/physiopathology , Retinopathy of Prematurity/physiopathology , Wet Macular Degeneration/physiopathology , Angiotensin-Converting Enzyme Inhibitors , Humans , Receptor, Angiotensin, Type 1 , Renin-Angiotensin System/physiology
11.
Exp Eye Res ; 176: 1-9, 2018 11.
Article in English | MEDLINE | ID: mdl-29944850

ABSTRACT

Hypertension is an independent risk factor for diabetic retinopathy, yet anti-hypertensive medications such as blockade of angiotensin II do not completely protect against vision-threatening vascular disease. We hypothesized that the potent vasoactive factor, endothelin (ET), is up-regulated in diabetic retinopathy and antagonism of the ET type A receptor (ETRA) or ET type B receptor (ETRB) ameliorates retinal vascular leakage independently of any blood pressure lowering effects. Spontaneously hypertensive rats (SHR) and their normotensive and genetic controls, Wistar Kyoto rats, were randomized to become diabetic or non-diabetic and studied for 8 weeks. Rats were further randomized to receive by intravitreal injection the ETRA antagonist, BQ123, the ETRB antagonist, BQ788, or vehicle, 5 days after the induction of streptozotocin diabetes and 4 weeks later. The treatments had no effect on systolic blood pressure which remained elevated in SHR. ET-1, ET-2, ETRA and ETRB were expressed in retina and retinal pigment epithelium (RPE)/choroid and increased by hypertension or diabetes. BQ123 reduced ET-1 and ET-2 expression in retina and RPE/choroid, while BQ788 had a similar effect but did not influence the mRNA levels of ET-1 in retina. Retinal vascular leakage and Müller cell stress as well as vascular endothelial growth factor (VEGF) expression in retina and RPE/choroid, were increased by hypertension or diabetes and there was an additive effect of these conditions. Treatment with BQ123 or BQ788 effectively reduced these events as well as the elevated levels of inflammatory factors in the retina. Our findings indicate that local ET systems exist in the retina and RPE/choroid that are up-regulated by hypertension and diabetes. The ability of locally delivered ET receptor antagonists to supress these overactive ET systems and reduce retinal vascular leakage and VEGF in the presence of hypertension indicate the potential of these approaches for the treatment of diabetic retinopathy.


Subject(s)
Antihypertensive Agents/therapeutic use , Diabetic Retinopathy/prevention & control , Endothelin A Receptor Antagonists/therapeutic use , Endothelin B Receptor Antagonists/therapeutic use , Ocular Hypertension/prevention & control , Animals , Blood Pressure/drug effects , Blood-Retinal Barrier/drug effects , Choroid/metabolism , Diabetes Mellitus, Experimental/prevention & control , Diabetic Retinopathy/metabolism , Endothelin A Receptor Antagonists/metabolism , Endothelin B Receptor Antagonists/metabolism , Endothelin-1/genetics , Endothelin-1/metabolism , Endothelin-2/genetics , Endothelin-2/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Intravitreal Injections , Ocular Hypertension/metabolism , Oligopeptides/therapeutic use , Peptides, Cyclic/therapeutic use , Piperidines/therapeutic use , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Streptozocin
12.
Clin Exp Ophthalmol ; 46(6): 652-660, 2018 08.
Article in English | MEDLINE | ID: mdl-29360265

ABSTRACT

BACKGROUND: Over-production of reactive oxygen species (ROS) and resulting oxidative stress contribute to retinal damage in vascular diseases that include diabetic retinopathy, retinopathy of prematurity and major retinal vessel occlusions. NADPH oxidase (Nox) proteins are professional ROS-generating enzymes, and therapeutic targeting in these diseases has strong appeal. Pharmacological inhibition of Nox4 reduces the severity of experimental retinal vasculopathy. We investigated the potential application of this drug approach in humans. METHODS: Differential Nox enzyme expression was studied by real-time-quantitative polymerase chain reaction in primary human retinal endothelial cell isolates and a characterized human retinal endothelial cell line. Oxidative stress was triggered chemically in endothelial cells, by treatment with dimethyloxalylglycine (DMOG; 100 µM); Nox4 and vascular endothelial growth factor (VEGFA) transcript were measured; and production of ROS was detected by 2',7'-dichlorofluorescein. DMOG-stimulated endothelial cells were treated with two Nox1/Nox4 inhibitors, GKT136901 and GKT137831; cell growth was monitored by DNA quantification, in addition to VEGFA transcript and ROS production. RESULTS: Nox4 (isoform Nox4A) was the predominant Nox enzyme expressed by human retinal endothelial cells. Treatment with DMOG significantly increased endothelial cell expression of Nox4 over 72 h, accompanied by ROS production and increased VEGFA expression. Treatment with GKT136901 or GKT137831 significantly reduced DMOG-induced ROS production and VEGFA expression by endothelial cells, and the inhibitory effect of DMOG on cell growth. CONCLUSIONS: Our findings in experiments on activated human retinal endothelial cells provide translational corroboration of studies in experimental models of retinal vasculopathy and support the therapeutic application of Nox4 inhibition by GKT136901 and GKT137831 in patients with retinal vascular diseases.


Subject(s)
Diabetic Retinopathy/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , NADPH Oxidase 1/genetics , Oxidative Stress , Retinal Vessels/pathology , Cell Proliferation , Cells, Cultured , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelial Cells/pathology , Humans , NADPH Oxidase 1/biosynthesis , RNA/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Vessels/metabolism
13.
Arterioscler Thromb Vasc Biol ; 36(6): 1186-96, 2016 06.
Article in English | MEDLINE | ID: mdl-27055905

ABSTRACT

OBJECTIVE: Although inhibitors of vascular endothelial growth factor (VEGF) provide benefit for the management of neovascular retinopathies, their use is limited to end-stage disease and some eyes are resistant. We hypothesized that retinoic acid-related orphan nuclear receptor γ (RORγ) and its downstream effector, interleukin (IL)-17A, upregulate VEGF and hence are important treatment targets for neovascular retinopathies. APPROACH AND RESULTS: Utilizing a model of oxygen-induced retinopathy, confocal microscopy and flow cytometry, we identified that retinal immunocompetent cells, microglia, express IL-17A. This was confirmed in primary cultures of rat retinal microglia, where hypoxia increased IL-17A protein as well as IL-17A, RORγ, and tumor necrosis factor-α mRNA, which were reduced by the RORγ inhibitor, digoxin, and the RORα/RORγ inverse agonist, SR1001. By contrast, retinal macroglial Müller cells and ganglion cells, key sources of VEGF in oxygen-induced retinopathy, did not produce IL-17A when exposed to hypoxia and IL-1ß. However, they expressed IL-17 receptors, and in response to IL-17A, secreted VEGF. This suggested that RORγ and IL-17A inhibition might attenuate neovascular retinopathy. Indeed, digoxin and SR1001 reduced retinal vaso-obliteration, neovascularization, and vascular leakage as well as VEGF and VEGF-related placental growth factor. Digoxin and SR1001 reduced microglial-derived IL-17A and Müller cell and ganglion cell damage. The importance of IL-17A in oxygen-induced retinopathy was confirmed by IL-17A neutralization reducing vasculopathy, VEGF, placental growth factor, tumor necrosis factor-α, microglial density and Müller cell, and ganglion cell injury. CONCLUSIONS: Our findings indicate that an RORγ/IL-17A axis influences VEGF production and neovascular retinopathy by mechanisms involving neuroglia. Inhibition of RORγ and IL-17A may have potential for the improved treatment of neovascular retinopathies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Digoxin/pharmacology , Interleukin-17/antagonists & inhibitors , Microglia/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Retina/drug effects , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Sulfonamides/pharmacology , Thiazoles/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Ependymoglial Cells/drug effects , Ependymoglial Cells/immunology , Ependymoglial Cells/metabolism , Hyperoxia/complications , Interleukin-17/genetics , Interleukin-17/metabolism , Mice, Inbred C57BL , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Placenta Growth Factor/metabolism , Rats, Sprague-Dawley , Retina/immunology , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism , Retinal Neovascularization/immunology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinopathy of Prematurity/immunology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Clin Sci (Lond) ; 130(15): 1375-87, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27005782

ABSTRACT

An imbalance in oxidative stress and antioxidant defense mechanisms contributes to the development of ischaemic retinopathies such as diabetic retinopathy and retinopathy of prematurity (ROP). Currently, the therapeutic utility of targeting key transcription factors to restore this imbalance remains to be determined. We postulated that dh404, an activator of nuclear factor erythroid-2 related factor 2 (Nrf2), the master regulator of oxidative stress responses, would attenuate retinal vasculopathy by mechanisms involving protection against oxidative stress-mediated damage to glia. Oxygen-induced retinopathy (OIR) was induced in neonatal C57BL/6J mice by exposure to hyperoxia (phase I) followed by room air (phase II). dh404 (1 mg/kg/every second day) reduced the vaso-obliteration of phase I OIR and neovascularization, vascular leakage and inflammation of phase II OIR. In phase I, the astrocytic template and vascular endothelial growth factor (VEGF) expression necessary for physiological angiogenesis are compromised resulting in vaso-obliteration. These events were attenuated by dh404 and related to dh404's ability to reduce the hyperoxia-induced increase in reactive oxygen species (ROS) and markers of cell damage as well as boost the Nrf2-responsive antioxidants in cultured astrocytes. In phase II, neovascularization and vascular leakage occurs following gliosis of Müller cells and their subsequent increased production of angiogenic factors. dh404 reduced Müller cell gliosis and vascular leakage in OIR as well as the hypoxia-induced increase in ROS and angiogenic factors with a concomitant increase in Nrf2-responsive antioxidants in cultured Müller cells. In conclusion, agents such as dh404 that reduce oxidative stress and promote antioxidant capacity offer a novel approach to lessen the vascular and glial cell damage that occurs in ischaemic retinopathies.


Subject(s)
Antioxidants/pharmacology , Ependymoglial Cells/drug effects , NF-E2-Related Factor 2/agonists , Oleanolic Acid/analogs & derivatives , Oxidative Stress/drug effects , Retina/drug effects , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Angiogenic Proteins/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Capillary Permeability/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Hyperoxia/complications , Inflammation Mediators/metabolism , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/pharmacology , Rats, Sprague-Dawley , Retina/metabolism , Retina/pathology , Retinal Neovascularization/etiology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Signal Transduction/drug effects
15.
J Neuroinflammation ; 12: 136, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26219952

ABSTRACT

BACKGROUND: Inflammation and the excess production of reactive oxygen species (ROS) contribute significantly to the pathogenesis of ischemic retinopathies such as diabetic retinopathy and retinopathy of prematurity. We hypothesized that GKT137831, a dual inhibitor of NADPH oxidases (NOX) 1 and NOX4, reduces inflammation in the ischemic retina by dampening the pro-inflammatory phenotype of retinal immune cells as well as macroglial Müller cells and neurons. METHODS: Ischemic retinopathy was induced in Sprague-Dawley rats by exposure to 80 % O2 cycled with 21 % O2 for 3 h per day from postnatal day (P) 0 to P11, followed by room air (P12 to P18). GKT137831 was administered P12 to P18 (60 mg/kg, subcutaneous) and comparisons were to room air controls. Retinal inflammation was examined by measuring leukocyte adherence to the retinal vasculature, ionized calcium-binding adaptor protein-1-positive microglia/macrophages, and the mRNA and protein levels of key inflammatory factors involved in retinal disease. Damage to Müller cells was evaluated by quantitating glial fibrillary acidic protein-positive cells and vascular leakage with an albumin ELISA. To verify the anti-inflammatory actions of GKT137831 on glia and neurons involved in ischemic retinopathy, primary cultures of rat retinal microglia, Müller cells, and ganglion cells were exposed to the in vitro counterpart of ischemia, hypoxia (0.5 %), and treated with GKT137831 for up to 72 h. ROS levels were evaluated with dihydroethidium and the protein and gene expression of inflammatory factors with quantitative PCR, ELISA, and a protein cytokine array. RESULTS: In the ischemic retina, GKT137831 reduced the increased leukocyte adherence to the vasculature, the pro-inflammatory phenotype of microglia and macroglia, the increased gene and protein expression of vascular endothelial growth factor, monocyte chemoattractant protein-1, and leukocyte adhesion molecules as well as vascular leakage. In all cultured cell types, GKT137831 reduced the hypoxia-induced increase in ROS levels and protein expression of various inflammatory mediators. CONCLUSIONS: NOX1/4 enzyme inhibition with GKT137831 has potent anti-inflammatory effects in the retina, indicating its potential as a treatment for a variety of vision-threatening retinopathies.


Subject(s)
NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Neuroglia/drug effects , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Retinitis/prevention & control , Animals , Cell Adhesion/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Hypoxia/complications , In Vitro Techniques , Intercellular Adhesion Molecule-1/metabolism , Ischemia/etiology , Ischemia/pathology , Ischemia/prevention & control , NADH, NADPH Oxidoreductases/drug effects , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Pyrazolones , Pyridones , Rats , Reactive Oxygen Species/metabolism , Retinitis/metabolism , Retinitis/pathology , Vascular Endothelial Growth Factor A/metabolism
16.
J Autoimmun ; 61: 9-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26027434

ABSTRACT

B cell-activating factor of the TNF family (BAFF) is an essential B cell survival factor. However, high levels of BAFF promote systemic lupus erythematosus (SLE) in mice and humans. Belimumab (anti-human BAFF) limits B cell survival and is approved for use in patients with SLE. Surprisingly, the efficacy of rituximab (anti-human CD20) in SLE remains controversial, despite depleting B cells more potently than belimumab. This raises the question of whether B cell depletion is really the mechanism of action of belimumab. In BAFF transgenic mice, SLE development is T cell-independent but relies on innate activation of B cells via TLRs, and TLR expression is modulated by the BAFF receptor TACI. Here, we show that loss of TACI on B cells protected against BAFF-mediated autoimmune manifestations while preserving B cells, suggesting that loss of BAFF signaling through TACI rather than loss of B cells may underpin the effect of belimumab in the clinic. Therefore, B cell-sparing blockade of TACI may offer a more specific and safer therapeutic alternative to broad B cell depletion in SLE.


Subject(s)
B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Animals , Autoantibodies/immunology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Flow Cytometry , Gene Expression/immunology , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism
17.
Exp Eye Res ; 136: 1-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25912997

ABSTRACT

Oxidative stress is an important contributor to glial and vascular cell damage in ischemic retinopathies. We hypothesized that ebselen via its ability to reduce reactive oxygen species (ROS) and augment nuclear factor-like 2 (Nrf2) anti-oxidants would attenuate hypoxia-induced damage to macroglial Müller cells and also lessen retinal vasculopathy. Primary cultures of rat Müller cells were exposed to normoxia (21% O2), hypoxia (0.5% O2) and ebselen (2.5 µM) for up to 72 h. Oxygen-induced retinopathy (OIR) was induced in C57BL/6J mice while control mice were housed in room air. Mice received vehicle (saline, 5% dimethyl sulfoxide) or ebselen (10 mg/kg) each day between postnatal days 6-18. In cultured Müller cells, flow cytometry for dihydroethidium revealed that ebselen reduced the hypoxia-induced increase in ROS levels, whilst increasing the expression of Nrf2-regulated anti-oxidant genes, heme oxygenase 1, glutathione peroxidase-1, NAD(P)H dehydrogenase quinone oxidoreductase 1 and glutamate-cysteine ligase. Moreover, in Müller cells, ebselen reduced the hypoxia-induced increase in protein levels of pro-angiogenic and pro-inflammatory factors including vascular endothelial growth factor, interleukin-6, monocyte chemoattractant-protein 1 and intercellular adhesion molecule-1, and the mRNA levels of glial fibrillary acidic protein (GFAP), a marker of Müller cell injury. Ebselen improved OIR by attenuating capillary vaso-obliteration and neovascularization and a concomitant reduction in Müller cell gliosis and GFAP. We conclude that ebselen protects against hypoxia-induced injury of retinal Müller cells and the microvasculature, which is linked to its ability to reduce oxidative stress, vascular damaging factors and inflammation. Agents such as ebselen may be potential treatments for retinopathies that feature oxidative stress-mediated damage to glia and the microvasculature.


Subject(s)
Antioxidants/pharmacology , Azoles/pharmacology , Ependymoglial Cells/drug effects , Gliosis/drug therapy , Hypoxia/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/drug effects , Retinal Degeneration/prevention & control , Animals , Animals, Newborn , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Ependymoglial Cells/metabolism , Flow Cytometry , Glial Fibrillary Acidic Protein , Gliosis/metabolism , Isoindoles , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Retinal Degeneration/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Superoxides/metabolism , Vascular System Injuries/prevention & control
18.
Arterioscler Thromb Vasc Biol ; 34(9): 2033-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25012132

ABSTRACT

OBJECTIVE: Neovascularization and vaso-obliteration are vision-threatening events that develop by interactions between retinal vascular and glial cells. A high-salt diet is causal in cardiovascular and renal disease, which is linked to modulation of the renin-angiotensin-aldosterone system. However, it is not known whether dietary salt influences retinal vasculopathy and if the renin-angiotensin-aldosterone system is involved. We examined whether a low-salt (LS) diet influenced vascular and glial cell injury and the renin-angiotensin-aldosterone system in ischemic retinopathy. APPROACH AND RESULTS: Pregnant Sprague Dawley rats were fed LS (0.03% NaCl) or normal salt (0.3% NaCl) diets, and ischemic retinopathy was induced in the offspring. An LS diet reduced retinal neovascularization and vaso-obliteration, the mRNA and protein levels of the angiogenic factors, vascular endothelial growth factor, and erythropoietin. Microglia, which influence vascular remodeling in ischemic retinopathy, were reduced by LS as was tumor necrosis factor-α. Macroglial Müller cells maintain the integrity of the blood-retinal barrier, and in ischemic retinopathy, LS reduced their gliosis and also vascular leakage. In retina, LS reduced mineralocorticoid receptor, angiotensin type 1 receptor, and renin mRNA levels, whereas, as expected, plasma levels of aldosterone and renin were increased. The aldosterone/mineralocorticoid receptor-sensitive epithelial sodium channel alpha (ENaCα), which is expressed in Müller cells, was increased in ischemic retinopathy and reduced by LS. In cultured Müller cells, high salt increased ENaCα, which was prevented by mineralocorticoid receptor and angiotensin type 1 receptor blockade. Conversely, LS reduced ENaCα, angiotensin type 1 receptor, and mineralocorticoid receptor expression. CONCLUSIONS: An LS diet reduced retinal vasculopathy, by modulating glial cell function and the retinal renin-angiotensin-aldosterone system.


Subject(s)
Diet, Sodium-Restricted , Epithelial Sodium Channels/physiology , Microglia/physiology , Renin-Angiotensin System/physiology , Retinal Neovascularization/diet therapy , Adaptor Protein Complex 1/analysis , Aldosterone/blood , Aldosterone/physiology , Animals , Animals, Newborn , Aquaporin 4/biosynthesis , Aquaporin 4/genetics , Body Weight , Cells, Cultured , Disease Models, Animal , Drinking Behavior , Ependymoglial Cells/chemistry , Ependymoglial Cells/pathology , Erythropoietin/analysis , Gliosis/etiology , Gliosis/physiopathology , Hematocrit , Ion Transport , Ischemia/physiopathology , Kidney Glomerulus/pathology , MAP Kinase Signaling System , Phosphorylation , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Retinal Neovascularization/physiopathology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity , Sodium/metabolism , Sodium Chloride, Dietary/adverse effects , Tumor Necrosis Factor-alpha/biosynthesis , Vascular Endothelial Growth Factor A/analysis
19.
Clin Exp Pharmacol Physiol ; 42(5): 537-48, 2015 May.
Article in English | MEDLINE | ID: mdl-25707593

ABSTRACT

Angiogenesis and inflammation are causative factors in the development of neovascular retinopathies. These processes involve the retinal endothelium and the retinal immune cells, microglia. The renin-angiotensin system contributes to retinal injury via the actions of the type 1 angiotensin receptor (AT1R). However, it has been suggested that prorenin, the initiator of the renin-angiotensin system cascade, influences retinal injury independently from the AT1R. We evaluated whether prorenin induced a pro-angiogenic and pro-inflammatory response in retinal endothelial cells and a pro-inflammatory phenotype in retinal microglia. Primary cultures of retinal endothelial cells and microglia were studied. Rat recombinant prorenin (2 nmol/L) stimulated the proliferation and tubulogenesis of retinal endothelial cells; it increased the levels of pro-angiogenic factors, vascular endothelial growth factor, angiopoietin-1, and tyrosine kinase with immunoglobulin and epidermal growth factor homology domains, and pro-inflammatory factors, intercellular adhesion molecule-1 and monocyte chemoattractant protein-1, relative to the controls. The messenger RNA levels of the (pro)renin receptor were also increased. These effects occurred in the presence of the AT1R blocker candesartan (10 µmol/L) and the renin inhibitor aliskiren (10 µmol/L). Microglia, which express the (pro)renin receptor, elicited an activated phenotype when exposed to prorenin, which was characterized by increased levels of intercellular adhesion molecule-1, monocyte chemoattractant protein-1, tumour necrosis factor-α, interleukin-6, and interleukin-1ß and by decreased levels of interleukin-10 and arginase-1 relative to controls. Candesartan did not influence the effects of prorenin on retinal microglia. In conclusion, prorenin has distinct pro-angiogenic and pro-inflammatory effects on retinal cells that are independent of the AT1R, indicating the potential importance of prorenin in retinopathy.


Subject(s)
Endothelial Cells/drug effects , Microglia/drug effects , Neovascularization, Physiologic/drug effects , Phenotype , Renin/pharmacology , Retina/cytology , Retina/drug effects , Animals , Cattle , Gene Expression Regulation/drug effects , Inflammation/metabolism , Inflammation/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Prorenin Receptor
20.
Nutr Diabetes ; 14(1): 46, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902253

ABSTRACT

BACKGROUND: Dietary-resistant starch is emerging as a potential therapeutic tool to limit the negative effects of diabetes on the kidneys. However, its metabolic and immunomodulatory effects have not yet been fully elucidated. METHODS: Six-week-old db/db mice were fed a diet containing 12.5% resistant starch or a control diet matched for equivalent regular starch for 10 weeks. db/m mice receiving the control diet were utilised as non-diabetic controls. Freshly collected kidneys were digested for flow cytometry analysis of immune cell populations. Kidney injury was determined by measuring albuminuria, histology, and immunohistochemistry. Portal vein plasma was collected for targeted analysis of microbially-derived metabolites. Intestinal histology and tight junction protein expression were assessed. RESULTS: Resistant starch limited the development of albuminuria in db/db mice. Diabetic db/db mice displayed a decline in portal vein plasma levels of acetate, propionate, and butyrate, which was increased with resistant starch supplementation. Diabetic db/db mice receiving resistant starch had a microbially-derived metabolite profile similar to that of non-diabetic db/m mice. The intestinal permeability markers lipopolysaccharide and lipopolysaccharide binding protein were increased in db/db mice consuming the control diet, which was not seen in db/db mice receiving resistant starch supplementation. Diabetes was associated with an increase in the kidney neutrophil population, neutrophil activation, number of C5aR1+ neutrophils, and urinary complement C5a excretion, all of which were reduced with resistant starch. These pro-inflammatory changes appear independent of fibrotic changes in the kidney. CONCLUSIONS: Resistant starch supplementation in diabetes promotes beneficial circulating microbially-derived metabolites and improves intestinal permeability, accompanied by a modulation in the inflammatory profile of the kidney including neutrophil infiltration, complement activation, and albuminuria. These findings indicate that resistant starch can regulate immune and inflammatory responses in the kidney and support the therapeutic potential of resistant starch supplementation in diabetes on kidney health.


Subject(s)
Albuminuria , Diabetic Nephropathies , Kidney , Neutrophil Infiltration , Animals , Mice , Kidney/metabolism , Male , Neutrophil Infiltration/drug effects , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/diet therapy , Resistant Starch/pharmacology , Gastrointestinal Microbiome/drug effects , Starch/pharmacology , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL