162 results on '"Joshua L. Dunaief"'
Search Results
2. Low ceruloplasmin levels exacerbate retinal degeneration in a hereditary hemochromatosis model
- Author
-
Brandon D. Anderson, Timothy Lee, Brent Bell, Ying Song, and Joshua L. Dunaief
- Subjects
ceruloplasmin ,iron ,retina ,hemochromatosis ,oxidative stress ,Medicine ,Pathology ,RB1-214 - Published
- 2023
- Full Text
- View/download PDF
3. A comparison of optophysiological biomarkers of photoreceptor stress and phototoxicity in BALB/cJ, B6(Cg)-Tyrc-2J/J, and C57Bl/6J mouse strains
- Author
-
Brent A. Bell, Charles Kaul, Joshua L. Dunaief, Joe G. Hollyfield, and Vera L. Bonilha
- Subjects
mice ,retina ,photoreceptors ,imaging ,photooxidation ,phototoxicity ,Medicine - Abstract
IntroductionOphthalmic imaging instruments, including the confocal scanning laser ophthalmoscope and spectral-domain optical coherence tomography system, originally intended for revealing ocular microstructures in the human eye, have been deployed by vision researchers to evaluate the eyes of numerous small and large animal species for more than two decades. In this study, we have used these two instruments to obtain imaging data sequentially from the retinas of three prominent, widely used experimental mouse models to document changes induced by two contrasting vivarium lighting conditions. Mice studied include albino BALB/cJ and B6(Cg)-Tyrc-2J/J and pigmented C57Bl/6J.MethodsMice were reared under dim light conditions until ~8 weeks of age where they underwent baseline imaging. Following, mice were returned to the dim vivarium or relocated to the top rack cage position in a standard vivarium. Mice were then followed for several months by ocular imaging to catalog the retinal dynamics as a function of long-term dim vs. elevated, standard vivarium lighting exposure levels.ResultsUpon exposure to elevated light levels, B6(Cg)-Tyrc-2J/J underwent similar changes as BALB/cJ in regard to photoreceptor outer segment shortening, photoreceptor layer proximal aspect hyperreflective changes, and the development of retinal infoldings and autofluorescent sub-retinal inflammatory monocyte infiltrate. Noteworthy, however, is that infoldings and infiltrate occurred at a slower rate of progression in B6(Cg)-Tyrc-2J/J vs. BALB/cJ. The photoreceptor outer nuclear layer thickness of BALB/cJ degenerated steadily following elevated light onset. In contrast, B6(Cg)-Tyrc-2J/J degeneration was unremarkable for many weeks before experiencing a noticeable change in the rate of degeneration that was concomitant with a plateau and decreasing trend in number of retinal infoldings and monocyte infiltrate. Pathological changes in C57Bl/6J mice were unremarkable for all imaging biomarkers assessed with exception to autofluorescent sub-retinal inflammatory monocyte infiltrate, which showed significant accumulation in dim vs. elevated light exposed mice following ~1 year of observation. These data were evaluated using Spearman’s correlation and Predictive Power Score matrices to determine the best imaging optophysiological biomarkers for indicating vivarium light stress and light-induced photoreceptor degeneration.DiscussionThis study suggests that changes in proximal aspect hyperreflectivity, outer segment shortening, retinal infoldings and autofluorescent sub-retinal inflammatory monocyte infiltrate are excellent indicators of light stress and light-induced degeneration in albino B6(Cg)-Tyrc-2J/J and BALB/cJ mouse strains.
- Published
- 2023
- Full Text
- View/download PDF
4. Retinal pigment epithelium-specific CLIC4 mutant is a mouse model of dry age-related macular degeneration
- Author
-
Jen-Zen Chuang, Nan Yang, Nobuyuki Nakajima, Wataru Otsu, Cheng Fu, Howard Hua Yang, Maxwell Ping Lee, Armaan Fazal Akbar, Tudor Constantin Badea, Ziqi Guo, Afnan Nuruzzaman, Kuo-Shun Hsu, Joshua L. Dunaief, and Ching-Hwa Sung
- Subjects
Science - Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness and is characterised by the accumulation of lipid deposits, called drusen. Here, the authors show that mice lacking chloride intracellular channel 4 in retinal pigment epithelium have defective lipid processing in the eye and pathological features mirroring human AMD, including drusen formation.
- Published
- 2022
- Full Text
- View/download PDF
5. Retina tissue validation of optical coherence tomography determined outer nuclear layer loss in FTLD-tau
- Author
-
Benjamin J. Kim, Vivian Lee, Edward B. Lee, Adrienne Saludades, John Q. Trojanowski, Joshua L. Dunaief, Murray Grossman, and David J. Irwin
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Alzheimer’s disease (AD) is associated with inner retina (nerve fiber and ganglion cell layers) thinning. In contrast, we have seen outer retina thinning driven by photoreceptor outer nuclear layer (ONL) thinning with antemortem optical coherence tomography (OCT) among patients considered to have a frontotemporal degeneration tauopathy (FTLD-Tau). Our objective was to determine if postmortem retinal tissue from FTLD-Tau patients demonstrates ONL loss observed antemortem on OCT. Two probable FTLD-Tau patients that were deeply phenotyped by clinical and genetic testing were imaged with OCT and followed to autopsy. Postmortem brain and retinal tissue were evaluated by a neuropathologist and ocular pathologist, respectively, masked to diagnosis. OCT findings were correlated with retinal histology. The two patients had autopsy-confirmed FTLD-Tau neuropathology and had antemortem OCT measurements showing ONL thinning (66.9 μm, patient #1; 74.9 μm, patient #2) below the 95% confidence interval of normal limits (75.1–120.7 μm) in our healthy control cohort. Postmortem, retinal tissue from both patients demonstrated loss of nuclei in the ONL, matching ONL loss visualized on antemortem OCT. Nuclei counts from each area of ONL loss (2 – 3 nuclei per column) seen in patient eyes were below the 95% confidence interval (4 – 8 nuclei per column for ONL) of 3 normal control retinas analyzed at the same location. Our evaluation of retinal tissue from FTLD-Tau patients confirms ONL loss seen antemortem by OCT. Continued investigation of ONL thinning as a biomarker that may distinguish FTLD-Tau from other dementias is warranted.
- Published
- 2021
- Full Text
- View/download PDF
6. Inflammatory adipose activates a nutritional immunity pathway leading to retinal dysfunction
- Author
-
Jacob K. Sterling, Bailey Baumann, Sierra Foshe, Andrew Voigt, Samyuktha Guttha, Ahab Alnemri, Sam J. McCright, Mingyao Li, Randy J. Zauhar, Sandra R. Montezuma, Rebecca J. Kapphahn, Venkata R.M. Chavali, David A. Hill, Deborah A. Ferrington, Dwight Stambolian, Robert F. Mullins, David Merrick, and Joshua L. Dunaief
- Subjects
CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1β (IL-1β). IL-1β upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.
- Published
- 2022
- Full Text
- View/download PDF
7. REV-ERBα regulates age-related and oxidative stress-induced degeneration in retinal pigment epithelium via NRF2
- Author
-
Shuo Huang, Chi-Hsiu Liu, Zhongxiao Wang, Zhongjie Fu, William R. Britton, Alexandra K. Blomfield, Theodore M. Kamenecka, Joshua L. Dunaief, Laura A. Solt, and Jing Chen
- Subjects
Retinal pigment epithelium ,Aging ,Age-related macular degeneration ,REV-ERBα ,Oxidative damage ,NRF2 ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Retinal pigment epithelium (RPE) dysfunction and atrophy occur in dry age-related macular degeneration (AMD), often leading to photoreceptor degeneration and vision loss. Accumulated oxidative stress during aging contributes to RPE dysfunction and degeneration. Here we show that the nuclear receptor REV-ERBα, a redox sensitive transcription factor, protects RPE from age-related degeneration and oxidative stress-induced damage. Genetic deficiency of REV-ERBα leads to accumulated oxidative stress, dysfunction and degeneration of RPE, and AMD-like ocular pathologies in aging mice. Loss of REV-ERBα exacerbates chemical-induced RPE damage, and pharmacological activation of REV-ERBα protects RPE from oxidative damage both in vivo and in vitro. REV-ERBα directly regulates transcription of nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream antioxidant enzymes superoxide dismutase 1 (SOD1) and catalase to counter oxidative damage. Moreover, aged mice with RPE specific knockout of REV-ERBα also exhibit accumulated oxidative stress and fundus and RPE pathologies. Together, our results suggest that REV-ERBα is a novel intrinsic protector of the RPE against age-dependent oxidative stress and a new molecular target for developing potential therapies to treat age-related retinal degeneration.
- Published
- 2022
- Full Text
- View/download PDF
8. Dexras1 Deletion and Iron Chelation Promote Neuroprotection in Experimental Optic Neuritis
- Author
-
Reas S. Khan, Bailey Baumann, Kimberly Dine, Ying Song, Joshua L. Dunaief, Sangwon F. Kim, and Kenneth S. Shindler
- Subjects
Medicine ,Science - Abstract
Abstract Dysregulation of iron metabolism, and resultant cytotoxicity, has been implicated in the pathogenesis of multiple sclerosis (MS) and other neurodegenerative processes. Iron accumulation promotes cytotoxicity through various mechanisms including oxidative stress and glutamate toxicity, and occurs in both MS patients and in the experimental autoimmune encephalomyelitis (EAE) model of MS. Divalent Metal Transporter1, a major iron importer in cells, is stimulated by signaling of Dexras1, a small G protein member of the Ras family. Dexras1 is activated by S-nitrosylation by nitric oxide (NO) produced by either inducible nitric oxide synthase in activated microglia/macrophages or neuronal nitric oxide synthase in neurons. Here we show Dexras1 exacerbates oxidative stress-induced neurodegeneration in experimental optic neuritis, an inflammatory demyelinating optic nerve condition that occurs in MS and EAE. Dexras1 deletion, as well as treatment with the iron chelator deferiprone, preserves vision and attenuates retinal ganglion cell (RGC) and axonal loss during EAE optic neuritis. These results suggest that iron entry triggered by NO-activated Dexras1 signaling is a potential mechanism of neuronal death in experimental optic neuritis. The current data suggest modulation of Dexras1 signaling and iron chelation are potential novel treatment strategies for optic neuritis and MS, and possibly other optic neuropathies as well.
- Published
- 2019
- Full Text
- View/download PDF
9. Mechanisms that minimize retinal impact of apolipoprotein E absence[S]
- Author
-
Aicha Saadane, Alexey Petrov, Natalia Mast, Nicole El-Darzi, Tung Dao, Ahab Alnemri, Ying Song, Joshua L. Dunaief, and Irina A. Pikuleva
- Subjects
cholesterol ,retina ,lipoproteins ,iron ,cytoskeleton ,vesicular traffic ,Biochemistry ,QD415-436 - Abstract
Apolipoprotein E (APOE) is a component of lipid-transporting particles and a recognition ligand for receptors, which bind these particles. The APOE isoform ε2 is a risk factor for age-related macular degeneration; nevertheless, APOE absence in humans and mice does not significantly affect the retina. We found that retinal cholesterol biosynthesis and the levels of retinal cholesterol were increased in Apoe−/− mice, whereas cholesterol elimination by metabolism was decreased. No focal cholesterol deposits were observed in the Apoe−/− retina. Retinal proteomics identified the most abundant cholesterol-related proteins in WT mice and revealed that, of these cholesterol-related proteins, only APOA4 had increased expression in the Apoe−/− retina. In addition, there were changes in retinal abundance of proteins involved in proinflammatory and antiinflammatory responses, cellular cytoskeleton maintenance, vesicular traffic, and retinal iron homeostasis. The data obtained indicate that when APOE is absent, particles containing APOA1, APOA4, and APOJ still transport cholesterol in the intraretinal space, but these particles are not taken up by retinal cells. Therefore, cholesterol biosynthesis inside retinal cells increase, whereas metabolism to oxysterols decreases to prevent cells from cholesterol depletion. These and other compensatory changes underlie only a minor retinal phenotype in Apoe−/− mice.
- Published
- 2018
- Full Text
- View/download PDF
10. GLP-1 Receptor Agonist NLY01 Reduces Retinal Inflammation and Neuron Death Secondary to Ocular Hypertension
- Author
-
Jacob K. Sterling, Modupe O. Adetunji, Samyuktha Guttha, Albert R. Bargoud, Katherine E. Uyhazi, Ahmara G. Ross, Joshua L. Dunaief, and Qi N. Cui
- Subjects
glaucoma ,neuroinflammation ,intraocular pressure ,A1 reactive astrocyte ,NLY01 ,retinal ganglion cell ,Biology (General) ,QH301-705.5 - Abstract
Summary: Glaucoma is the leading cause of irreversible blindness and is characterized by the death of retinal ganglion cells (RGCs). Recent studies have implicated pro-inflammatory microglia, macrophages, and A1 astrocytes in the pathogenesis of neurodegenerative diseases. The role of pro-inflammatory, neurotoxic A1 astrocytes in glaucoma is just beginning to be explored. Using a mouse model of glaucoma, we demonstrate that ocular hypertension is sufficient to trigger production of C1q, interleukin-1α (IL-1α), and tumor necrosis factor α (TNF-α), three cytokines necessary and sufficient to drive the formation of A1 astrocytes. Upregulation of these cytokines occurs first in CD11b+ CD11c+ cells followed by CD11b+ CD11c− cells. Ablation of this pathway, by either genetic deletions of C1qa, IL-1α, and TNF-α, or treatment with glucagon-like peptide-1 receptor agonist NLY01, reduces A1 astrocyte transformation and RGC death. Together, these results highlight a neuroinflammatory mechanism of glaucomatous neurodegeneration that can be therapeutically targeted by NLY01 administration.
- Published
- 2020
- Full Text
- View/download PDF
11. Ferrous but not ferric iron sulfate kills photoreceptors and induces photoreceptor-dependent RPE autofluorescence
- Author
-
Wanting Shu, Bailey H. Baumann, Ying Song, Yingrui Liu, Xingwei Wu, and Joshua L. Dunaief
- Subjects
Oxidative stress ,Iron ,Photoreceptor ,Retinal pigment epithelium (RPE) ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Iron has been implicated in the pathogenesis of retinal degenerative diseases, including ocular siderosis. However, the mechanisms of iron-induced retinal toxicity are incompletely understood. Previous work shows that intravitreal injection of Fe2+ leads to photoreceptor (PR) oxidative stress, resulting in PR death within 14 days, and cones are more susceptible than rods to iron-induced oxidative damage. In order to further investigate the mechanism of intravitreal iron-induced retinal toxicity and shed light on mechanisms of iron-induced retinopathy in other mouse models, Fe2+, Fe3+, or saline were injected into the vitreous of adult wild-type mice. Pre-treatment with Ferrostatin-1 was used to investigate whether iron-induced retinal toxicity resulted from ferroptosis. Color and autofluorescence in vivo retinal imaging and optical coherence tomography were performed on day 2 and day 7 post-injection. Eyes were collected for quantitative PCR and Western analysis on day 1 and for immunofluorescence on both day 2 and 7. In vivo imaging and immunofluorescence revealed that Fe2+, but not Fe3+, induced PR oxidative damage and autofluorescence on day 2, resulting in PR death and retinal pigment epithelial cell (RPE) autofluorescence on day 7. Quantitative PCR and Western analysis on day 1 indicated that both Fe2+ and Fe3+ induced iron accumulation in the retina. However, only Fe2+ elevated levels of oxidative stress markers and components of ferroptosis in the retina, and killed PRs. Ferrostatin-1 failed to protect the retina from Fe2+-induced oxidative damage. To investigate the mechanism of Fe2+-induced RPE autofluorescence, rd10 mutant mice aged 6 weeks, with almost total loss of PRs, were given intravitreal Fe2+ or Fe3+ injections: neither induced RPE autofluorescence. This result suggests Fe2+-induced RPE autofluorescence in wild-type mice resulted from phagocytosed, oxidized outer segments. Together these data suggest that intraretinal Fe2+ causes PR oxidative stress, leading to PR death and RPE autofluorescence.
- Published
- 2020
- Full Text
- View/download PDF
12. Bull's eye maculopathy associated with hereditary hemochromatosis
- Author
-
Kellyn N. Bellsmith, Joshua L. Dunaief, Paul Yang, Mark E. Pennesi, Ellen Davis, Holly Hofkamp, and Brandon J. Lujan
- Subjects
Retinopathy ,Iron ,Copper ,Ferritin ,Optical coherence tomography ,Retinal pigment epithelium ,Ophthalmology ,RE1-994 - Abstract
Purpose: To report a case of bull's eye maculopathy, a novel finding in a patient with iron overload secondary to hereditary hemochromatosis with a homozygous mutation of the HFE gene. Observations: A 39-year-old man with recently diagnosed hereditary hemochromatosis undergoing treatment by serial phlebotomy presented with bilateral progressive blurry vision and recent onset of photopsias and headaches. Fundus examination revealed a symmetric bull's eye maculopathy with photoreceptor loss and retinal pigment epithelium transmission defects in the area of speckled hyper- and hypo-pigmentation by multimodal imaging. Full field and multifocal electroretinograms demonstrated generalized rod and cone dysfunction with some central preservation of waveforms. Further systemic work-up revealed low ceruloplasmin, mildly decreased serum copper and zinc levels, and low urinary copper. The patient underwent testing for inherited retinal dystrophies, but was not found to have any known pathogenic gene mutations. His ferritin levels normalized with serial phlebotomy and his retinopathy did not appear to progress over 6 months with normalization of his iron levels. Conclusions and Importance: We report a case of bull's eye maculopathy in a patient with hereditary hemochromatosis with no previous exposure to iron chelators and no known inherited retinal dystrophy. Ocular involvement in hereditary hemochromatosis is relatively rare. In this case, the patient's low serum ceruloplasmin is thought to have increased the amount of redox-active ferrous iron and potentiated retinal iron toxicity resulting in the observed retinopathy. To the authors' knowledge, this is a potentially novel ocular manifestation of hereditary hemochromatosis.
- Published
- 2020
- Full Text
- View/download PDF
13. Severe Iron Metabolism Defects in Mice With Double Knockout of the Multicopper Ferroxidases Hephaestin and CeruloplasminSummary
- Author
-
Brie K. Fuqua, Yan Lu, David M. Frazer, Deepak Darshan, Sarah J. Wilkins, Linda Dunn, Alex V. Loguinov, Scott C. Kogan, Pavle Matak, Huijun Chen, Joshua L. Dunaief, Chris D. Vulpe, and Gregory J. Anderson
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: Multicopper ferroxidases (MCFs) facilitate intestinal iron absorption and systemic iron recycling, likely by a mechanism involving the oxidization of Fe2+ from the iron exporter ferroportin 1 for delivery to the circulating Fe3+ carrier transferrin. Hephaestin (HEPH), the only MCF known to be expressed in enterocytes, aids in the basolateral transfer of dietary iron to the blood. Mice lacking HEPH in the whole body (Heph-/-) or intestine alone (Hephint/int) exhibit defects in dietary iron absorption but still survive and grow. Circulating ceruloplasmin (CP) is the only other known MCF likely to interact with enterocytes. Our aim was to assess the effects of combined deletion of HEPH and CP on intestinal iron absorption and homeostasis in mice. Methods: Mice lacking both HEPH and CP (Heph-/-Cp-/-) and mice with whole-body knockout of CP and intestine-specific deletion of HEPH (Hephint/intCp-/-) were generated and phenotyped. Results: Heph-/-Cp-/- mice were severely anemic and had low serum iron, but they exhibited marked iron loading in duodenal enterocytes, the liver, heart, pancreas, and other tissues. Hephint/intCp-/- mice were moderately anemic (similar to Cp-/- mice) but were iron loaded only in the duodenum and liver, as in Hephint/int and Cp-/- mice, respectively. Both double knockout models absorbed iron in radiolabeled intestinal iron absorption studies, but the iron was inappropriately distributed, with an abnormally high percentage retained in the liver. Conclusions: These studies indicate that HEPH and CP, and likely MCFs in general, are not essential for intestinal iron absorption but are required for proper systemic iron distribution. They also point to important extra-intestinal roles for HEPH in maintaining whole-body iron homeostasis. Keywords: Iron Deficiency Anemia, Iron Overload, Intestinal Iron Absorption, Non-Transferrin Bound Iron
- Published
- 2018
- Full Text
- View/download PDF
14. Persistent and Progressive Outer Retina Thinning in Frontotemporal Degeneration
- Author
-
Benjamin J. Kim, Murray Grossman, Delu Song, Samantha Saludades, Wei Pan, Sophia Dominguez-Perez, Joshua L. Dunaief, Tomas S. Aleman, Gui-Shuang Ying, and David J. Irwin
- Subjects
frontotemporal degeneration ,optical coherence tomography ,retina ,tauopathy ,progressive supranuclear palsy ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
ObjectiveWhile Alzheimer’s disease is associated with inner retina thinning measured by spectral-domain optical coherence tomography (SD-OCT), our previous cross-sectional study suggested outer retina thinning in frontotemporal degeneration (FTD) patients compared to controls without neurodegenerative disease; we sought to evaluate longitudinal changes of this potential biomarker.MethodsSD-OCT retinal layer thicknesses were measured at baseline and after 1–2 years. Clinical criteria, genetic analysis, and a cerebrospinal fluid biomarker (total tau: β-amyloid) to exclude likely underlying Alzheimer’s disease pathology were used to define a subgroup of predicted molecular pathology (i.e., tauopathy). Retinal layer thicknesses and rates of change in all FTD patients (n = 16 patients, 30 eyes) and the tauopathy subgroup (n = 9 patients,16 eyes) were compared to controls (n = 30 controls, 47 eyes) using a generalized linear model accounting for inter-eye correlation and adjusting for age, sex, and race. Correlations between retinal layer thicknesses and Mini-Mental State Examinations (MMSE) were assessed.ResultsCompared to controls, returning FTD patients (143 vs. 130 μm, p = 0.005) and the tauopathy subgroup (143 vs. 128 μm, p = 0.03) had thinner outer retinas but similar inner layer thicknesses. Compared to controls, the outer retina thinning rate was not significant for all FTD patients (p = 0.34), but was significant for the tauopathy subgroup (−3.9 vs. 0.4 μm/year, p = 0.03). Outer retina thickness change correlated with MMSE change in FTD patients (Spearman rho = 0.60, p = 0.02) and the tauopathy subgroup (rho = 0.73, p = 0.04).ConclusionOur finding of FTD outer retina thinning persists and longitudinally correlates with disease progression. These findings were especially seen in probable tauopathy patients, which showed progressive outer retina thinning.
- Published
- 2019
- Full Text
- View/download PDF
15. Iron Toxicity in the Retina Requires Alu RNA and the NLRP3 Inflammasome
- Author
-
Bradley D. Gelfand, Charles B. Wright, Younghee Kim, Tetsuhiro Yasuma, Reo Yasuma, Shengjian Li, Benjamin J. Fowler, Ana Bastos-Carvalho, Nagaraj Kerur, Annette Uittenbogaard, Youn Seon Han, Dingyuan Lou, Mark E. Kleinman, W. Hayes McDonald, Gabriel Núñez, Philippe Georgel, Joshua L. Dunaief, and Jayakrishna Ambati
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Excess iron induces tissue damage and is implicated in age-related macular degeneration (AMD). Iron toxicity is widely attributed to hydroxyl radical formation through Fenton’s reaction. We report that excess iron, but not other Fenton catalytic metals, induces activation of the NLRP3 inflammasome, a pathway also implicated in AMD. Additionally, iron-induced degeneration of the retinal pigmented epithelium (RPE) is suppressed in mice lacking inflammasome components caspase-1/11 or Nlrp3 or by inhibition of caspase-1. Iron overload increases abundance of RNAs transcribed from short interspersed nuclear elements (SINEs): Alu RNAs and the rodent equivalent B1 and B2 RNAs, which are inflammasome agonists. Targeting Alu or B2 RNA prevents iron-induced inflammasome activation and RPE degeneration. Iron-induced SINE RNA accumulation is due to suppression of DICER1 via sequestration of the co-factor poly(C)-binding protein 2 (PCBP2). These findings reveal an unexpected mechanism of iron toxicity, with implications for AMD and neurodegenerative diseases associated with excess iron.
- Published
- 2015
- Full Text
- View/download PDF
16. Potential Treatment of Retinal Diseases with Iron Chelators
- Author
-
Wanting Shu and Joshua L. Dunaief
- Subjects
chelation ,iron ,retina ,age-related macular degeneration (AMD) ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Iron is essential for life, while excess iron can be toxic. Iron generates hydroxyl radical, which is the most reactive free radical, causing oxidative stress. Since iron is absorbed through the diet but not excreted from the body, it accumulates with age in tissues, including the retina, consequently leading to age-related toxicity. This accumulation is further promoted by inflammation. Hereditary diseases such as aceruloplasminemia, Friedreich’s ataxia, pantothenate kinase-associated neurodegeneration, and posterior column ataxia with retinitis pigmentosa involve retinal degeneration associated with iron dysregulation. In addition to hereditary causes, dietary or parenteral iron supplementation has been recently reported to elevate iron levels in the retinal pigment epithelium (RPE) and promote retinal degeneration. Ocular siderosis from intraocular foreign bodies or subretinal hemorrhage can also lead to retinopathy. Evidence from mice and humans suggests that iron toxicity may contribute to age-related macular degeneration pathogenesis. Iron chelators can protect photoreceptors and RPE in various mouse models. The therapeutic potential for iron chelators is under investigation.
- Published
- 2018
- Full Text
- View/download PDF
17. Retinal iron homeostasis in health and disease
- Author
-
Delu eSong and Joshua L. Dunaief
- Subjects
Ceruloplasmin ,Iron ,Oxidative Stress ,Retina ,ferritin ,Age-related degeneration ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Iron is essential for life, but excess iron can be toxic. As a potent free radical creator, iron generates hydroxyl radicals leading to significant oxidative stress. Since iron is not excreted from the body, it accumulates with age in tissues, including the retina, predisposing to age-related oxidative insult. Both hereditary and acquired retinal diseases are associated with increased iron levels. For example, retinal degenerations have been found in hereditary iron overload disorders, like aceruloplasminemia, Friedreich’s ataxia, and pantothenate kinase-associated neurodegeneration. Similarly, mice with targeted mutation of the iron exporter ceruloplasmin and its homolog hephaestin showed age-related retinal iron accumulation and retinal degeneration with features resembling human age-related macular degeneration (AMD). Post mortem AMD eyes have increased levels of iron in retina compared to age-matched healthy donors. Iron accumulation in AMD is likely to result, in part, from inflammation, hypoxia, and oxidative stress, all of which can cause iron dysregulation. Fortunately, it has been demonstrated by in vitro and in vivo studies that iron in the retinal pigment epithelium and retina is chelatable. Iron chelation protects photoreceptors and retinal pigment epithelial cells (RPE) in a variety of mouse models. This has therapeutic potential for diminishing iron-induced oxidative damage to prevent or treat AMD.
- Published
- 2013
- Full Text
- View/download PDF
18. Systemic Medication Use and the Incidence and Growth of Geographic Atrophy in the Comparison of Age-related Macular Degeneration Treatments Trials (CATT)
- Author
-
Delu, Song, Peiying, Hua, Brian L, VanderBeek, Joshua L, Dunaief, Juan E, Grunwald, Ebenezer, Daniel, Maureen G, Maguire, Daniel F, Martin, and Gui-Shuang, Ying
- Published
- 2020
- Full Text
- View/download PDF
19. Chronic migraine reversal and prevention with the LIFE diet: a nutrient dense whole food plant-based diet (WFPBD)
- Author
-
Brittany Perzia, Joshua L. Dunaief, and David M. Dunaief
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Diet, Vegetarian ,Migraine Disorders ,Headache ,Complete blood count ,Plant based ,General Medicine ,Nutrients ,Middle Aged ,medicine.disease ,Diet ,Nutrient density ,Chronic Migraine ,Migraine ,medicine ,Humans ,Whole food ,Migraine treatment ,Headaches ,medicine.symptom ,business - Abstract
We report a case of a 60-year-old man who struggled with frequent migraines for 12.5 years, which were refractory to all conventional therapies. Six months before initial consultation, these migraines become chronic. The patient was then advised to follow the Low Inflammatory Foods Everyday (LIFE) diet, a nutrient-dense, dark green leafy vegetable-rich, whole food plant-based diet. Within 2 months, his headache frequency declined from 18 to 24 headache days per month to 1, and he discontinued his preventive and abortive migraine medications. After 3 months, the patient had no headaches. These results far exceed the goal of migraine treatment with medication, which is to reduce migraine frequency by >50% per month. In addition, the results were durable; this patient has been migraine-free for 7.5 years. Serum beta-carotene more than tripled after the patient started the LIFE diet, consistent with its high content of dark green leafy vegetables. Weight, high-sensitivity C-reactive protein (hsCRP), complete blood count (CBC), hydration status, sodium and other electrolytes remained constant throughout the study.
- Published
- 2023
20. Impaired ABCA1/ABCG1-mediated lipid efflux in the mouse retinal pigment epithelium (RPE) leads to retinal degeneration
- Author
-
Federica Storti, Katrin Klee, Vyara Todorova, Regula Steiner, Alaa Othman, Saskia van der Velde-Visser, Marijana Samardzija, Isabelle Meneau, Maya Barben, Duygu Karademir, Valda Pauzuolyte, Sanford L Boye, Frank Blaser, Christoph Ullmer, Joshua L Dunaief, Thorsten Hornemann, Lucia Rohrer, Anneke den Hollander, Arnold von Eckardstein, Jürgen Fingerle, Cyrille Maugeais, and Christian Grimm
- Subjects
ABCA1 ,retinal degeneration ,retinal pigment epithelium ,lipid efflux ,age-related macular degeneration ,ABCG1 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Age-related macular degeneration (AMD) is a progressive disease of the retinal pigment epithelium (RPE) and the retina leading to loss of central vision. Polymorphisms in genes involved in lipid metabolism, including the ATP-binding cassette transporter A1 (ABCA1), have been associated with AMD risk. However, the significance of retinal lipid handling for AMD pathogenesis remains elusive. Here, we study the contribution of lipid efflux in the RPE by generating a mouse model lacking ABCA1 and its partner ABCG1 specifically in this layer. Mutant mice show lipid accumulation in the RPE, reduced RPE and retinal function, retinal inflammation and RPE/photoreceptor degeneration. Data from human cell lines indicate that the ABCA1 AMD risk-conferring allele decreases ABCA1 expression, identifying the potential molecular cause that underlies the genetic risk for AMD. Our results highlight the essential homeostatic role for lipid efflux in the RPE and suggest a pathogenic contribution of reduced ABCA1 function to AMD.
- Published
- 2019
- Full Text
- View/download PDF
21. Retina tissue validation of optical coherence tomography determined outer nuclear layer loss in FTLD-tau
- Author
-
Vivian S. Lee, Edward B. Lee, Joshua L. Dunaief, Murray Grossman, David J. Irwin, John Q. Trojanowski, Adrienne Saludades, and Benjamin J. Kim
- Subjects
Male ,medicine.medical_specialty ,genetic structures ,Nerve fiber ,Autopsy ,Cell Count ,tau Proteins ,Case Report ,Neuropathology ,Retina ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Optical coherence tomography ,Ophthalmology ,mental disorders ,medicine ,Humans ,Outer nuclear layer ,RC346-429 ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Brain ,Retinal ,Middle Aged ,Retinal Photoreceptor Cell Outer Segment ,eye diseases ,Ganglion ,medicine.anatomical_structure ,chemistry ,Female ,Neurology (clinical) ,Supranuclear Palsy, Progressive ,sense organs ,Neurology. Diseases of the nervous system ,Frontotemporal Lobar Degeneration ,business ,Biomarkers ,Tomography, Optical Coherence - Abstract
Alzheimer’s disease (AD) is associated with inner retina (nerve fiber and ganglion cell layers) thinning. In contrast, we have seen outer retina thinning driven by photoreceptor outer nuclear layer (ONL) thinning with antemortem optical coherence tomography (OCT) among patients considered to have a frontotemporal degeneration tauopathy (FTLD-Tau). Our objective was to determine if postmortem retinal tissue from FTLD-Tau patients demonstrates ONL loss observed antemortem on OCT. Two probable FTLD-Tau patients that were deeply phenotyped by clinical and genetic testing were imaged with OCT and followed to autopsy. Postmortem brain and retinal tissue were evaluated by a neuropathologist and ocular pathologist, respectively, masked to diagnosis. OCT findings were correlated with retinal histology. The two patients had autopsy-confirmed FTLD-Tau neuropathology and had antemortem OCT measurements showing ONL thinning (66.9 μm, patient #1; 74.9 μm, patient #2) below the 95% confidence interval of normal limits (75.1–120.7 μm) in our healthy control cohort. Postmortem, retinal tissue from both patients demonstrated loss of nuclei in the ONL, matching ONL loss visualized on antemortem OCT. Nuclei counts from each area of ONL loss (2 – 3 nuclei per column) seen in patient eyes were below the 95% confidence interval (4 – 8 nuclei per column for ONL) of 3 normal control retinas analyzed at the same location. Our evaluation of retinal tissue from FTLD-Tau patients confirms ONL loss seen antemortem by OCT. Continued investigation of ONL thinning as a biomarker that may distinguish FTLD-Tau from other dementias is warranted.
- Published
- 2021
22. Once-Daily Low Inflammatory Foods Everyday (LIFE) Smoothie or the Full LIFE Diet Lowers C-Reactive Protein and Raises Plasma Beta-Carotene in 7 Days
- Author
-
David M. Dunaief, Joshua L. Dunaief, Brittany Perzia, and Gui-Shuang Ying
- Subjects
Original Researchs ,medicine.medical_specialty ,Longitudinal study ,biology ,business.industry ,Health Policy ,C-reactive protein ,Public Health, Environmental and Occupational Health ,Medicine (miscellaneous) ,030204 cardiovascular system & hematology ,Systemic inflammation ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Increased risk ,beta-Carotene ,Internal medicine ,medicine ,biology.protein ,030212 general & internal medicine ,medicine.symptom ,Once daily ,business ,Everyday life - Abstract
Serum C-reactive protein (CRP), a marker of systemic inflammation, is associated with increased risk for numerous inflammation-driven chronic diseases. A prior longitudinal study showed that the Low Inflammatory Foods Everyday (LIFE) diet, which is rich in dark green leafy vegetables (DGLV), lowered CRP over a mean follow-up period of 6 months. In this retrospective study, we investigate whether patients who consume the LIFE diet or their regular diet plus one component of the LIFE diet (LIFE smoothie), experience reductions in high-sensitivity CRP (hsCRP) in 7 days. Sixteen patients in a community practice met inclusion criteria. Patient compliance was assessed by patient interviews and measurements of beta-carotene, which is abundant in DGLV. Following the interventions, CRP decreased in both the LIFE diet (−0.47 mg/L, P = .02) and smoothie groups (−1.2 mg/L, P = .04). No statistically significant difference in reduction was observed between groups ( P = .18). Plasma beta-carotene increased in both groups (+23.2, P = .02; +20.6, P = .006, respectively). These findings suggest that the LIFE diet or a regular American diet supplemented with the LIFE smoothie may quickly reduce systemic inflammation and the risk of many chronic diseases.
- Published
- 2022
23. Conditional knockout of hephaestin in the neural retina disrupts retinal iron homeostasis
- Author
-
Kevin R. Zhang, Bailey Baumann, Ying Song, Jacob Sterling, Elizabeth A. Erler, Samyuktha Guttha, Zbynek Kozmik, and Joshua L. Dunaief
- Subjects
Mice, Knockout ,Cellular and Molecular Neuroscience ,Ophthalmology ,Macular Degeneration ,Mice ,Iron ,Animals ,Ceruloplasmin ,Homeostasis ,Membrane Proteins ,Sensory Systems ,Article ,Retina - Abstract
Iron accumulation has been implicated in degenerative retinal diseases. It can catalyze the production of damaging reactive oxygen species. Previous work has demonstrated iron accumulation in multiple retinal diseases, including age-related macular degeneration and diabetic retinopathy. In mice, systemic knockout of the ferroxidases ceruloplasmin (Cp) and hephaestin (Heph), which oxidize iron, results in retinal iron accumulation and iron-induced degeneration. To determine the role of Heph in the retina, we generated a neural retina-specific Heph knockout on a background of systemic Cp knockout. This resulted in elevated neural retina iron. Conversely, retinal ganglion cells had elevated transferrin receptor and decreased ferritin, suggesting diminished iron levels. The retinal degeneration observed in systemic Cp(−/−), Heph(−/−) mice did not occur. These findings indicate that Heph has a local role in regulating neural retina iron homeostasis, but also suggest that preserved Heph function in either the RPE or systemically mitigates the degeneration phenotype observed in the systemic Cp(−/−), Heph(−/−) mice.
- Published
- 2022
24. Orally Administered Alpha Lipoic Acid as a Treatment for Geographic Atrophy
- Author
-
Yafeng Li, Apurva Patel, Gui-Shuang Ying, Ebenezer Daniel, Albert O. Edwards, Juan E. Grunwald, Karen M. Gehrs, Paul Hahn, Benjamin J. Kim, Allan A. Hunter, Itzhak Nissim, Rahul N. Khurana, Joshua L. Dunaief, Alexander J. Brucker, Maureen G. Maguire, and Maxwell Pistilli
- Subjects
0303 health sciences ,medicine.medical_specialty ,Visual acuity ,genetic structures ,business.industry ,Phases of clinical research ,Macular degeneration ,Fundus (eye) ,medicine.disease ,Placebo ,Confidence interval ,law.invention ,03 medical and health sciences ,Ophthalmology ,0302 clinical medicine ,Randomized controlled trial ,law ,030221 ophthalmology & optometry ,Medicine ,sense organs ,medicine.symptom ,business ,Adverse effect ,030304 developmental biology - Abstract
Purpose Alpha lipoic acid (ALA) is a nutraceutical and potent antioxidant that has shown efficacy in the retina light damage mouse model and in humans for multiple sclerosis. Our objective was to evaluate the efficacy and safety of oral ALA for the treatment of geographic atrophy (GA). Design Randomized, controlled, double-masked, multicenter phase 2 clinical trial of ALA versus placebo. Participants Participants with unilateral or bilateral GA from age-related macular degeneration. Methods Participants were randomized to 1200 mg daily of ALA or placebo. Fundus autofluorescence, fundus color photography, and spectral-domain OCT were conducted and best-corrected visual acuity (BCVA) was obtained at baseline and every 6 months through month 18. Main Outcome Measures Annual rate of change over 18 months in square root-transformed area of GA in study eyes as measured on fundus autofluorescence. Secondary outcomes included the number of adverse events (AEs), change in BCVA, and annual rate of change in area of GA measured on color photographs. Results Fifty-three participants (mean age, 80 years) were randomized (April 2016–August 2017). Twenty-seven participants (37 eyes) were in the placebo group, and 26 participants (36 eyes) were in the ALA group. Unadjusted mean (standard error) annual change in GA area was 0.28 (0.02) mm and 0.31 (0.02) mm for the placebo and ALA groups, respectively (difference, 0.04 mm; 95% confidence interval [CI], –0.03 to 0.11 mm; P = 0.30). Adjusting for baseline GA area, number of GA lesions, and presence of subfoveal GA, the mean annual change in GA area was 0.27 (0.04) mm and 0.32 (0.05) mm for the placebo and ALA groups, respectively (difference, 0.05 mm; 95% CI, –0.02 to 0.12 mm; P = 0.14). At 18 months, the percent of eyes losing 15 letters or more of BCVA was 22% (8 of 36) and 14% (5 of 36) in the placebo and ALA groups, respectively (P = 0.54). No difference was found in the percentage of participants with nonserious AEs (P = 0.96) or serious AEs (P = 0.28) between the placebo and ALA groups. Conclusions Results do not support ALA having beneficial effects on GA or BCVA. This trial design may be useful for other GA repurposing drug trials.
- Published
- 2020
25. Minimal effect of conditional ferroportin KO in the neural retina implicates ferrous iron in retinal iron overload and degeneration
- Author
-
Yingrui Liu, Bailey Baumann, Ying Song, Kevin Zhang, Jacob K. Sterling, Samira Lakhal-Littleton, Zbynek Kozmik, Guanfang Su, and Joshua L. Dunaief
- Subjects
Mice, Knockout ,Cellular and Molecular Neuroscience ,Ophthalmology ,Mice ,Iron Overload ,Iron ,Animals ,Ceruloplasmin ,Cation Transport Proteins ,Sensory Systems ,Article ,Retina - Abstract
Iron-induced oxidative stress can cause or exacerbate retinal degenerative diseases. Retinal iron overload has been reported in several mouse disease models with systemic or neural retina-specific knockout (KO) of homologous ferroxidases ceruloplasmin (Cp) and hephaestin (Heph). Cp and Heph can potentiate ferroportin (Fpn) mediated cellular iron export. Here, we used retina-specific Fpn KO mice to test the hypothesis that retinal iron overload in Cp/Heph DKO mice is caused by impaired iron export from neurons and glia. Surprisingly, there was no indication of retinal iron overload in retina-specific Fpn KO mice: the mRNA levels of transferrin receptor in the retina were not altered at 7-10-months age. Consistent with this, levels and localization of ferritin light chain were unchanged. To "stress the system", we injected iron intraperitoneally into Fpn KO mice with or without Cp KO. Only mice with both retina-specific Fpn KO and Cp KO had modestly elevated retinal iron levels. These results suggest that impaired iron export through Fpn is not sufficient to explain the retinal iron overload in Cp/Heph DKO mice. An increase in the levels of retinal ferrous iron caused by the absence of these ferroxidases, followed by uptake into cells by ferrous iron importers, is most likely necessary.
- Published
- 2022
26. Deuterated docosahexaenoic acid protects against oxidative stress and geographic atrophy-like retinal degeneration in a mouse model with iron overload
- Author
-
Yingrui Liu, Brent A. Bell, Ying Song, Kevin Zhang, Brandon Anderson, Paul H. Axelsen, Whitney Bohannan, Martin‐Paul Agbaga, Hui Gyu Park, Genevieve James, J. Thomas Brenna, Karsten Schmidt, Joshua L. Dunaief, and Mikhail S. Shchepinov
- Subjects
Aging ,Iron Overload ,Docosahexaenoic Acids ,Iron ,Retinal Degeneration ,Cell Biology ,Retinal Pigment Epithelium ,Disease Models, Animal ,Macular Degeneration ,Mice ,Oxidative Stress ,Geographic Atrophy ,Animals ,Humans - Abstract
Oxidative stress plays a central role in age-related macular degeneration (AMD). Iron, a potent generator of hydroxyl radicals through the Fenton reaction, has been implicated in AMD. One easily oxidized molecule is docosahexaenoic acid (DHA), the most abundant polyunsaturated fatty acid in photoreceptor membranes. Oxidation of DHA produces toxic oxidation products including carboxyethylpyrrole (CEP) adducts, which are increased in the retinas of AMD patients. In this study, we hypothesized that deuterium substitution on the bis-allylic sites of DHA in photoreceptor membranes could prevent iron-induced retinal degeneration by inhibiting oxidative stress and lipid peroxidation. Mice were fed with either DHA deuterated at the oxidation-prone positions (D-DHA) or control natural DHA and then given an intravitreal injection of iron or control saline. Orally administered D-DHA caused a dose-dependent increase in D-DHA levels in the neural retina and retinal pigment epithelium (RPE) as measured by mass spectrometry. At 1 week after iron injection, D-DHA provided nearly complete protection against iron-induced retinal autofluorescence and retinal degeneration, as determined by in vivo imaging, electroretinography, and histology. Iron injection resulted in carboxyethylpyrrole conjugate immunoreactivity in photoreceptors and RPE in mice fed with natural DHA but not D-DHA. Quantitative PCR results were consistent with iron-induced oxidative stress, inflammation, and retinal cell death in mice fed with natural DHA but not D-DHA. Taken together, our findings suggest that DHA oxidation is central to the pathogenesis of iron-induced retinal degeneration. They also provide preclinical evidence that dosing with D-DHA could be a viable therapeutic strategy for retinal diseases involving oxidative stress.
- Published
- 2022
27. Rising Plasma Beta-Carotene Is Associated With Diminishing C-Reactive Protein in Patients Consuming a Dark Green Leafy Vegetable–Rich, Low Inflammatory Foods Everyday (LIFE) Diet
- Author
-
David M. Dunaief, Gui-Shuang Ying, Hannah Schultz, and Joshua L. Dunaief
- Subjects
biology ,business.industry ,Health Policy ,C-reactive protein ,Public Health, Environmental and Occupational Health ,Medicine (miscellaneous) ,Inflammation ,030204 cardiovascular system & hematology ,03 medical and health sciences ,0302 clinical medicine ,beta-Carotene ,Chart review ,biology.protein ,Green Leafy Vegetable ,Medicine ,In patient ,030212 general & internal medicine ,Food science ,medicine.symptom ,business ,Original Research - Abstract
Chronic inflammation contributes to a number of chronic diseases and can be assessed with C-reactive protein (CRP). In this longitudinal retrospective chart review, we investigate whether patients intensively counseled to eat a specific diet high in dark green leafy vegetables, and thus high beta-carotene, have reductions in plasma high-sensitivity CRP (hsCRP). We term this the Low Inflammatory Foods Everyday (LIFE) diet. Forty-three patients in a community practice instructed to eat the LIFE diet met inclusion criteria. The CRP levels were measured at least twice over the course of up to a year. Adherence to the diet was objectively assessed by measurement of plasma beta-carotene, which is abundant in dark green leafy vegetables, and subjectively by serial interviews. The change in beta-carotene was inversely correlated with change in CRP ( r = −0.68, P < .0001). Additionally, patients subjectively classified as adherent had higher beta-carotene ( P < .0001) and lower CRP ( P = .002) as compared with patients who were classified as nonadherent. These longitudinal findings suggest that adherence to the LIFE diet leads to increased beta-carotene and decreased CRP. Thus, this type of diet may reduce risk or severity of chronic diseases involving inflammation.
- Published
- 2019
28. Heavy lipids protect against heavy metals
- Author
-
Joshua L, Dunaief
- Subjects
Macular Degeneration ,Oxidative Stress ,Aging ,Metals, Heavy ,Humans ,Cell Biology ,Lipids - Published
- 2022
29. Decision letter: Aging is associated with increased brain iron through cortex-derived hepcidin expression
- Author
-
Joshua L Dunaief
- Published
- 2021
30. Intraocular iron injection induces oxidative stress followed by elements of geographic atrophy and sympathetic ophthalmia
- Author
-
Maura Poli, Janet R. Sparrow, Michelle Guo, Hye J. Kim, Jacob Sterling, Joshua L. Dunaief, Brent A. Bell, Y. Liu, Guanfang Su, Ying Song, Aditya M Rao, Benjamin J. Kim, and Kevin Zhang
- Subjects
Male ,Aging ,genetic structures ,Iron ,photoreceptor cells ,Inflammation ,Retinal Pigment Epithelium ,Biology ,medicine.disease_cause ,Ferric Compounds ,Lipofuscin ,Pathogenesis ,chemistry.chemical_compound ,Mice ,Geographic Atrophy ,medicine ,Animals ,lipofuscin ,Retina ,Sympathetic ophthalmia ,Optical Imaging ,Retinal ,lipid peroxidation ,Cell Biology ,medicine.disease ,Molecular biology ,eye diseases ,Mice, Inbred C57BL ,Quaternary Ammonium Compounds ,Disease Models, Animal ,Oxidative Stress ,Choroidal neovascularization ,medicine.anatomical_structure ,chemistry ,Ophthalmia, Sympathetic ,Original Article ,sense organs ,medicine.symptom ,Injections, Intraocular ,Oxidative stress - Abstract
Iron has been implicated in the pathogenesis of age‐related retinal diseases, including age‐related macular degeneration (AMD). Previous work showed that intravitreal (IVT) injection of iron induces acute photoreceptor death, lipid peroxidation, and autofluorescence (AF). Herein, we extend this work, finding surprising chronic features of the model: geographic atrophy and sympathetic ophthalmia. We provide new mechanistic insights derived from focal AF in the photoreceptors, quantification of bisretinoids, and localization of carboxyethyl pyrrole, an oxidized adduct of docosahexaenoic acid associated with AMD. In mice given IVT ferric ammonium citrate (FAC), RPE died in patches that slowly expanded at their borders, like human geographic atrophy. There was green AF in the photoreceptor ellipsoid, a mitochondria‐rich region, 4 h after injection, followed later by gold AF in rod outer segments, RPE and subretinal myeloid cells. The green AF signature is consistent with flavin adenine dinucleotide, while measured increases in the bisretinoid all‐trans‐retinal dimer are consistent with the gold AF. FAC induced formation carboxyethyl pyrrole accumulation first in photoreceptors, then in RPE and myeloid cells. Quantitative PCR on neural retina and RPE indicated antioxidant upregulation and inflammation. Unexpectedly, reminiscent of sympathetic ophthalmia, autofluorescent myeloid cells containing abundant iron infiltrated the saline‐injected fellow eyes only if the contralateral eye had received IVT FAC. These findings provide mechanistic insights into the potential toxicity caused by AMD‐associated retinal iron accumulation. The mouse model will be useful for testing antioxidants, iron chelators, ferroptosis inhibitors, anti‐inflammatory medications, and choroidal neovascularization inhibitors., Intravitreal injection of iron induces photoreceptor oxidative stress, resulting in increased autofluorescence, lipid peroxidation, and myeloid cell infiltration associated with retinal degeneration, geographic atrophy, and choroidal neovascularization.
- Published
- 2021
31. Myeloid cells in retinal and brain degeneration
- Author
-
Turner D. Schwartz, Joshua L. Dunaief, Michelle Guo, and Qi N. Cui
- Subjects
Parkinson's disease ,Central nervous system ,Biochemistry ,Neuroprotection ,Retina ,Article ,chemistry.chemical_compound ,Retinitis pigmentosa ,medicine ,Animals ,Myeloid Cells ,Molecular Biology ,Inflammation ,Microglia ,business.industry ,Brain ,Retinal ,Neurodegenerative Diseases ,Cell Biology ,Macular degeneration ,medicine.disease ,medicine.anatomical_structure ,chemistry ,business ,Neuroscience ,Retinitis Pigmentosa - Abstract
Retinal inflammation underlies multiple prevalent ocular and neurological diseases. Similar inflammatory processes are observed in glaucomatous optic neuropathy, age-related macular degeneration, retinitis pigmentosa, posterior uveitis, Alzheimer’s disease, and Parkinson’s disease. In particular, human and animal studies have demonstrated the important role microglia/macrophages play in initiating and maintaining a pro-inflammatory environment in degenerative processes impacting vision. On the other hand, microglia have also been shown to have a protective role in multiple central nervous system diseases. Identifying the mechanisms underlying cell dysfunction and death is the first step towards developing novel therapeutics for these diseases impacting the central nervous system. In addition to reviewing recent key studies defining important mediators of retinal inflammation, with an emphasis on translational studies that bridge this research from bench to bedside, we also highlight a promising therapeutic class of medications, the glucagon-like peptide-1 receptor agonists. Finally, we propose areas where additional research is necessary to identify mechanisms that can be modulated to shift the balance from a neurotoxic to a neuroprotective retinal environment.
- Published
- 2021
32. Multi-copper ferroxidase deficiency leads to iron accumulation and oxidative damage in astrocytes and oligodendrocytes
- Author
-
Gregory J. Anderson, Min Chen, Guohao Liu, Joshua L. Dunaief, Tianqi Shen, Chris D. Vulpe, Jiashuo Zheng, Ruiwei Jiang, Mengxia Chen, Nady Braidy, Huijun Chen, Zaitunamu Maimaitiming, Shunli Liu, and Zheng Chen
- Subjects
0301 basic medicine ,Hephaestin ,Iron ,Central nervous system ,lcsh:Medicine ,medicine.disease_cause ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,lcsh:Science ,Gene ,Mice, Knockout ,Messenger RNA ,Multidisciplinary ,biology ,Chemistry ,lcsh:R ,Ceruloplasmin ,Membrane Proteins ,Glial biology ,Transmembrane protein ,Cell biology ,Mice, Inbred C57BL ,Oligodendroglia ,Oxidative Stress ,Metabolism ,030104 developmental biology ,medicine.anatomical_structure ,Cerebral cortex ,Astrocytes ,biology.protein ,lcsh:Q ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Accumulation of iron has been associated with the pathobiology of various disorders of the central nervous system. Our previous work has shown that hephaestin (Heph) and ceruloplasmin (Cp) double knockout (KO) mice induced iron accumulation in multiple brain regions and that this was paralleled by increased oxidative damage and deficits in cognition and memory. In this study, we enriched astrocytes and oligodendrocytes from the cerebral cortex of neonatal wild-type (WT), Heph KO and Cp KO mice. We demonstrated that Heph is highly expressed in oligodendrocytes, while Cp is mainly expressed in astrocytes. Iron efflux was impaired in Cp KO astrocytes and Heph KO oligodendrocytes and was associated with increased oxidative stress. The expression of Heph, Cp, and other iron-related genes was examined in astrocytes and oligodendrocytes both with and without iron treatment. Interestingly, we found that the expression of the mRNA encoding ferroportin 1, a transmembrane protein that cooperates with CP and HEPH to export iron from cells, was positively correlated with Cp expression in astrocytes, and with Heph expression in oligodendrocytes. Our findings collectively demonstrate that HEPH and CP are important for the prevention of glial iron accumulation and thus may be protective against oxidative damage.
- Published
- 2019
33. Differential contribution of C5aR and C5b-9 pathways to renal thrombic microangiopathy and macrovascular thrombosis in mice carrying an atypical hemolytic syndrome–related factor H mutation
- Author
-
Damodar Gullipalli, Daisuke Ito, Wen-Chao Song, Sayaka Sato, Hangsoo Kim, Lin Zhou, Joshua L. Dunaief, Madhu Golla, Yoshiyasu Ueda, Matthew Palmer, Takashi Miwa, and Delu Song
- Subjects
Male ,0301 basic medicine ,Thrombotic microangiopathy ,Kidney Glomerulus ,030232 urology & nephrology ,Mice, Transgenic ,Complement Membrane Attack Complex ,urologic and male genital diseases ,Article ,C5a receptor ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Atypical hemolytic uremic syndrome ,medicine ,Animals ,Humans ,Point Mutation ,Complement Activation ,Receptor, Anaphylatoxin C5a ,Atypical Hemolytic Uremic Syndrome ,business.industry ,Microangiopathy ,Antibodies, Monoclonal ,Thrombosis ,C5 Deficiency ,medicine.disease ,female genital diseases and pregnancy complications ,Complement C6 ,Complement system ,Disease Models, Animal ,Microscopy, Electron ,030104 developmental biology ,Nephrology ,Complement Factor H ,Mutation ,Immunology ,Female ,business - Abstract
Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6−/− and FHR/R C9−/− mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.
- Published
- 2019
34. Five-Year Follow-up of Nonfibrotic Scars in the Comparison of Age-Related Macular Degeneration Treatments Trials
- Author
-
Maureen G. Maguire, Ebenezer Daniel, Juan E. Grunwald, Glenn J. Jaffe, Joshua L. Dunaief, Cynthia A. Toth, Daniel F. Martin, Wei Pan, Gui-Shuang Ying, Frederick L. Ferris, and Benjamin J. Kim
- Subjects
medicine.medical_specialty ,Visual acuity ,genetic structures ,Scars ,03 medical and health sciences ,0302 clinical medicine ,Atrophy ,Ophthalmology ,Medicine ,Prospective cohort study ,030304 developmental biology ,0303 health sciences ,medicine.diagnostic_test ,business.industry ,Macular degeneration ,Fluorescein angiography ,medicine.disease ,eye diseases ,Choroidal neovascularization ,030221 ophthalmology & optometry ,sense organs ,medicine.symptom ,Ranibizumab ,business ,medicine.drug - Abstract
Purpose To describe changes in visual acuity (VA) and macular morphologic features at 5 years in eyes with nonfibrotic scar (NFS) identified at 1 year in the Comparison of Age-Related Macular Degeneration Treatments Trials (CATT). Design Prospective cohort study within a randomized clinical trial. Participants Participants in CATT. Methods Participants assigned to ranibizumab or bevacizumab and to 1 of 3 dosing regimens were released from the clinical trial protocol after 2 years and recalled at 5 years. Nonfibrotic scar was identified on color images at year 1 as flat, small, well-circumscribed areas of pigmentation with varying degrees of central hypopigmentation without exposure of underlying choroidal vessels at the site of baseline choroidal neovascularization. Follow-up images were assessed for changes in and around NFS. Main Outcome Measures Pigmentation changes, VA, development of fibrotic scar (FS), nongeographic atrophy (NGA), geographic atrophy (GA), retinal fluid on OCT, and fluorescein leakage. Results Among 474 eyes with images obtained at 1, 2, and 5 years, 39 (8.2%) showed NFS at 1 year with a mean VA of 80 letters (Snellen equivalent, 20/25). Among these eyes, FS developed in 5% at 2 years and 28% at 5 years. Nongeographic atrophy was observed in 34%, 47%, and 65% of eyes at 1, 2, and 5 years, respectively. Geographic atrophy developed in 5% of eyes at 2 years and 21% at 5 years. Among eyes with NFS, FS, or no scar at 1 year, mean VA at 5 years was 73 letters (20/32), 48 letters (20/100), and 62 letters (20/63), respectively. At 5 years, NFS eyes demonstrated less GA, less intraretinal fluid, more subretinal fluid, and less subretinal pigment epithelium fluid (all P 0.50). The proportion of eyes with fluid on OCT also did not change (P = 0.36). Subretinal hyperreflective material disappeared by 5 years in 40% of eyes with NFS. Conclusions These results indicate that, on average, eyes with NFS after anti-VEGF treatment have good VA not only at 1 and 2 years, but also through 5 years.
- Published
- 2019
35. Comparative localization of cystathionine beta synthases and cystathionine gamma lyase in canine, non-human primate and human retina
- Author
-
Gustavo D. Aguirre, Joshua L. Dunaief, Raghavi Sudharsan, Alireza Badiei, and Evelyn Santana
- Subjects
Primates ,0301 basic medicine ,Retinal degeneration ,Blotting, Western ,Nerve fiber layer ,Cystathionine beta-Synthase ,Outer plexiform layer ,Retina ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Dogs ,0302 clinical medicine ,medicine ,Animals ,Laser capture microdissection ,biology ,Chemistry ,Cystathionine gamma-Lyase ,Retinal ,medicine.disease ,Immunohistochemistry ,Cystathionine beta synthase ,Sensory Systems ,Cell biology ,Blot ,Ophthalmology ,030104 developmental biology ,medicine.anatomical_structure ,030221 ophthalmology & optometry ,biology.protein ,sense organs - Abstract
Chronic exposure of the retina to light and high concentrations of polyunsaturated fatty acid in photoreceptor cells make this tissue susceptible to oxidative damage. As retinal degenerative diseases are associated with photoreceptor degeneration, the antioxidant activity of both hydrogen sulfide (H(2)S) and glutathione (GSH) may play an important role in ameliorating disease progression. H(2)S production is driven by cystathionine-γ-lyase (CSE) and cystathionine β–synthase (CBS), the key enzymes that also drive transsulfuration pathway (TSP) necessary for GSH production. As it is currently unclear whether localized production of either H(2)S or GSH contributes to retinal homeostasis, we undertook a comparative analysis of CBS and CSE expression in canine, non-human primates (NHP) and human retinas to determine if these antioxidants could play a regulatory role in age-related or disease-associated retinal degeneration. Retinas from normal dogs, NHPs and humans were used for the study. Laser capture microdissection (LCM) was performed to isolate individual layers of the canine retina and analyze CBS and CSE gene expression by qRT-PCR. Immunohistochemistry and western blotting were performed for CBS and CSE labeling and protein expression in dog, NHP, and human retina, respectively. Using qRT-PCR, western blot, and immunohistochemistry (IHC), we showed that CBS and CSE are expressed in the canine, NHP, and human retina. IHC results from canine retina demonstrated increased expression levels of CBS but not CSE with post-developmental aging. IHC results also showed non-overlapping localization of both proteins with CBS presenting in rods, amacrine, horizontal, and nerve fiber cell layers while CSE was expressed by RPE, cones and Mϋller cells. Finally, we demonstrated that these enzymes localized to all three layers of canine, NHP and human retina: photoreceptors, outer plexiform layer (OPL) and notably in the ganglion cells layer/nerve fiber layer (GCL/NFL). QRT-PCR performed using RNA extracted from tissues isolated from these cell layers using laser captured microdissection (LCM) confirmed that each of CBS and CSE are expressed equally in these three layers. Together, these findings reveal that CSE and CBS are expressed in the retina, thereby supporting further studies to determine the role of H(2)S and these proteins in oxidative stress and apoptosis in retinal degenerative diseases.
- Published
- 2019
36. Complement Factor H Mutation W1206R Causes Retinal Thrombosis and Ischemic Retinopathy in Mice
- Author
-
Yoshiyasu Ueda, Rupak Bhuyan, Takashi Miwa, Lin Zhou, Damodar Gullipali, Hangsoo Kim, Albert Bargoud, Delu Song, Imran Mohammed, Hannah Schultz, Joshua L. Dunaief, Ying Song, and Wen-Chao Song
- Subjects
Retinal degeneration ,Pathology ,medicine.medical_specialty ,Thrombotic microangiopathy ,Retinal Pigment Epithelium ,Article ,Pathology and Forensic Medicine ,Mice ,chemistry.chemical_compound ,Retinal Diseases ,Ischemia ,medicine ,Animals ,Retinal thinning ,Mice, Knockout ,Retinal Vascular Occlusion ,Retina ,Retinal pigment epithelium ,Neovascularization, Pathologic ,business.industry ,Thrombosis ,Retinal ,Macular degeneration ,medicine.disease ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,Complement Factor H ,Mutation ,embryonic structures ,business - Abstract
© 2019 American Society for Investigative Pathology Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FH R/R ) share features with human atypical hemolytic uremic syndrome. Herein, we report that FH R/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FH R/R mice. Optical coherence tomography imaging of FH R/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FH R/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FH R/R mice, consistent with membrane attack complex–mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FH R/R mice. This FH R/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.
- Published
- 2019
37. Reduction in Ferritin Concentrationsamong Patients Consuming a Dark-Green Leafy Vegetable–Rich, Low Inflammatory Foods Everyday (LIFE) Diet
- Author
-
Brittany M, Perzia, Gui-Shuang, Ying, Joshua L, Dunaief, and David M, Dunaief
- Subjects
Nutrition and Dietetics ,Medicine (miscellaneous) ,Food Science - Abstract
Ferritin is an iron-containing protein and acute-phase reactant, which may be elevated due to systemic iron overload or inflammation. Various diseases are associated with excess iron, but therapeutic iron chelation is suboptimal. Prior studies suggest that several plant phytochemicals possess iron-chelating properties, indicating that a plant-based diet may benefit patients with iron overload.The aim was to investigate whether patients who consume a nutrient-dense, dark-green leafy vegetable-rich diet, called the Low Inflammatory Foods Everyday (LIFE) diet, experience reductions in ferritin concentrations.This was a retrospective study in which patients were intensively counseled to follow the LIFE diet. Compliance was assessed by patient interviews and serum B-carotene measurements. Primary outcomes included changes in ferritin, B-carotene, and C-reactive protein (CRP). Patients with elevated CRP concentrations at baseline were excluded in order to separate the impact of inflammation from iron overload on ferritin concentrations. Premenopausal women, who lose iron from menstruation, were also excluded.Thirty-two patients met the inclusion criteria. The median follow-up was 183 d. Following the dietary intervention, ferritin decreased (-81 μg/L,The LIFE diet, or similar dark-green leafy vegetable-rich, whole-food plant-based diets, may benefit patients with disorders of iron overload and iron-induced oxidative stress.
- Published
- 2022
38. The multicopper ferroxidase hephaestin enhances intestinal iron absorption in mice.
- Author
-
Brie K Fuqua, Yan Lu, Deepak Darshan, David M Frazer, Sarah J Wilkins, Natalie Wolkow, Austin G Bell, JoAnn Hsu, Catherine C Yu, Huijun Chen, Joshua L Dunaief, Gregory J Anderson, and Chris D Vulpe
- Subjects
Medicine ,Science - Abstract
Hephaestin is a vertebrate multicopper ferroxidase important for the transfer of dietary iron from intestinal cells to the blood. Hephaestin is mutated in the sex-linked anemia mouse, resulting in iron deficiency. However, sex-linked anemia mice still retain some hephaestin ferroxidase activity. They survive, breed, and their anemia improves with age. To gain a better understanding of the role of hephaestin in iron homeostasis, we used the Cre-lox system to generate knockout mouse models with whole body or intestine-specific (Villin promoter) ablation of hephaestin. Both types of mice were viable, indicating that hephaestin is not essential and that other mechanisms, multicopper ferroxidase-dependent or not, must compensate for hephaestin deficiency. The knockout strains, however, both developed a microcytic, hypochromic anemia, suggesting severe iron deficiency and confirming that hephaestin plays an important role in body iron acquisition. Consistent with this, the knockout mice accumulated iron in duodenal enterocytes and had reduced intestinal iron absorption. In addition, the similarities of the phenotypes of the whole body and intestine-specific hephaestin knockout mice clarify the important role of hephaestin specifically in intestinal enterocytes in maintaining whole body iron homeostasis. These mouse models will serve as valuable tools to study the role of hephaestin and associated proteins in iron transport in the small intestine and other tissues.
- Published
- 2014
- Full Text
- View/download PDF
39. Mechanisms that minimize retinal impact of apolipoprotein E absence
- Author
-
Irina A. Pikuleva, Nicole El-Darzi, Ahab Alnemri, Tung Dao, Ying Song, Alexey M. Petrov, Natalia Mast, Joshua L. Dunaief, and Aicha Saadane
- Subjects
Male ,0301 basic medicine ,Gene isoform ,Apolipoprotein E ,Iron ,vesicular traffic ,QD415-436 ,Biochemistry ,Retina ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Apolipoproteins E ,0302 clinical medicine ,Endocrinology ,Tandem Mass Spectrometry ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Receptor ,Apolipoproteins A ,Cytoskeleton ,Research Articles ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Cholesterol ,Calcium-Binding Proteins ,Microfilament Proteins ,Retinal ,Cell Biology ,Metabolism ,Immunohistochemistry ,Cell biology ,lipoproteins ,Mice, Inbred C57BL ,Clusterin ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030221 ophthalmology & optometry ,Female ,lipids (amino acids, peptides, and proteins) - Abstract
Apolipoprotein E (APOE) is a component of lipid-transporting particles and a recognition ligand for receptors, which bind these particles. The APOE isoform ε2 is a risk factor for age-related macular degeneration; nevertheless, APOE absence in humans and mice does not significantly affect the retina. We found that retinal cholesterol biosynthesis and the levels of retinal cholesterol were increased in Apoe(−/−) mice, whereas cholesterol elimination by metabolism was decreased. No focal cholesterol deposits were observed in the Apoe(−/−) retina. Retinal proteomics identified the most abundant cholesterol-related proteins in WT mice and revealed that, of these cholesterol-related proteins, only APOA4 had increased expression in the Apoe(−/−) retina. In addition, there were changes in retinal abundance of proteins involved in proinflammatory and antiinflammatory responses, cellular cytoskeleton maintenance, vesicular traffic, and retinal iron homeostasis. The data obtained indicate that when APOE is absent, particles containing APOA1, APOA4, and APOJ still transport cholesterol in the intraretinal space, but these particles are not taken up by retinal cells. Therefore, cholesterol biosynthesis inside retinal cells increase, whereas metabolism to oxysterols decreases to prevent cells from cholesterol depletion. These and other compensatory changes underlie only a minor retinal phenotype in Apoe(−/−) mice.
- Published
- 2018
40. Glucagon-like peptide 1 receptor agonist use is associated with reduced risk for glaucoma
- Author
-
Jacob Sterling, Qi N. Cui, Peiying Hua, Joshua L. Dunaief, and Brian L VanderBeek
- Subjects
Oncology ,Agonist ,medicine.medical_specialty ,Intraocular pressure ,business.industry ,medicine.drug_class ,Type 2 Diabetes Mellitus ,Glaucoma ,Retrospective cohort study ,medicine.disease ,Retinal ganglion ,Sensory Systems ,Cellular and Molecular Neuroscience ,Ophthalmology ,Internal medicine ,Diabetes mellitus ,Medicine ,business ,Glucagon-like peptide 1 receptor - Abstract
Background/aimsGlucagon-like peptide-1 receptor (GLP-1R) agonists regulate blood glucose and are commonly used to treat type 2 diabetes mellitus. Recent work showed that treatment with the GLP-1R agonist NLY01 decreased retinal neuroinflammation and glial activation to rescue retinal ganglion cells in a mouse model of glaucoma. In this study, we used an insurance claims database (Clinformatics Data Mart) to examine whether GLP-1R agonist exposure impacts glaucoma risk.MethodsA retrospective cohort of patients who initiated a new GLP-1R agonist was 1:3 age, gender, race, classes of active diabetes medications and year of index date matched to patients who initiated a different class of oral diabetic medication. Inverse probability of treatment weighting (IPTW) was used within a multivariable Cox proportional hazard regression model to test the association between GLP-1R agonist exposure and a new diagnosis of primary open-angle glaucoma, glaucoma suspect or low-tension glaucoma.ResultsCohorts were comprised of 1961 new users of GLP-1R agonists matched to 4371 unexposed controls. After IPTW, all variables were balanced (standard mean deviation ConclusionsGLP-1R agonist use was associated with a statistically significant hazard reduction for a new diagnosis of glaucoma. Our findings support further investigations into the use of GLP-1R agonists in glaucoma prevention.
- Published
- 2021
41. Exposure to glucagon-like peptide 1 receptor (GLP-1R) agonists reduces glaucoma risk
- Author
-
Brian L VanderBeek, Jacob Sterling, Joshua L. Dunaief, Qi N. Cui, and Peiying Hua
- Subjects
Agonist ,medicine.medical_specialty ,Open angle glaucoma ,medicine.drug_class ,business.industry ,Semaglutide ,Glaucoma ,medicine.disease ,Retinal ganglion ,Albiglutide ,Low Tension Glaucoma ,Internal medicine ,medicine ,Dulaglutide ,business ,medicine.drug - Abstract
ImportanceGlucagon-like peptide-1 receptor (GLP-1R) agonists regulate blood glucose and are commonly used to treat Type II Diabetes Mellitus. Recent work has shown that treatment with the novel GLP-1R agonist, NLY01, decreased retinal neuroinflammation and glial activation to rescue retinal ganglion cells in an animal model of glaucoma.ObjectiveIn this study, we used an insurance claims database to examine whether GLP-1R agonist exposure impacts glaucoma risk.Design, Setting, and ParticipantsA retrospective cohort of adult patients who initiated a new GLP-1R agonist (i.e., exenatide, liraglutide, albiglutide, dulaglutide, semaglutide, or lixisenatide) was 1:3 age, gender, race, active diabetes medication classes, and year of index date matched to a cohort of patients who initiated a different class of oral diabetic medication during their time in the database. Exclusion occurred for ExposureGlucagon-like peptide 1 receptor agonist.Main Outcomes and MeasuresNew diagnosis of primary open angle glaucoma, glaucoma suspect, or low tension glaucoma.ResultsCohorts were comprised of 1,961 new users of GLP-1R agonists matched to 4,371 unexposed controls. After IPTW, age was the only covariate imbalanced (SMD >0.1) between cohorts. Ten new diagnoses of glaucoma (0.51%) were present in the GLP-1R agonist cohort compared to 58 (1.33%) in the unexposed controls. After adjustment, GLP-1R exposure conferred a reduced hazard of 0.54 (95%CI: 0.35-0.85, P =0.007), suggesting that GLP-1R agonists reduce the risk for glaucoma.Conclusions and RelevanceGLP-1R agonist use was associated with a statistically significant hazard reduction for a new glaucoma diagnosis. Our findings support further investigations into the use of GLP-1R agonists in glaucoma prevention.
- Published
- 2021
42. A vicious cycle of bisretinoid formation and oxidation relevant to recessive Stargardt disease
- Author
-
Hye Jin Kim, Joshua L. Dunaief, Keiko Ueda, Janet R. Sparrow, Diego Montenegro, Jin Zhao, and Kevin Zhang
- Subjects
0301 basic medicine ,Retinal degeneration ,retina ,retinal pigment epithelium ,ONL, outer nuclear layer ,RPE, retinal pigment epithelial cells ,Biochemistry ,Fenton reaction ,chemistry.chemical_compound ,Mice ,iron ,Stargardt Disease ,AMD, age-related macular degeneration ,ONH, optic nerve head ,lipofuscin ,chemistry.chemical_classification ,Mice, Knockout ,biology ,Cell biology ,medicine.anatomical_structure ,Retinaldehyde ,light ,Oxidation-Reduction ,Visual phototransduction ,Research Article ,cis-trans-Isomerases ,Genes, Recessive ,Lipofuscin ,bisretinoid ,03 medical and health sciences ,Retinoids ,Hepcidin ,medicine ,Animals ,Molecular Biology ,Reactive oxygen species ,ERG, electroretinography ,Retinal pigment epithelium ,030102 biochemistry & molecular biology ,Cell Biology ,STGD1, Stargardt Disease 1 ,medicine.disease ,eye diseases ,Ferritin ,030104 developmental biology ,chemistry ,ABCA4, ATP-binding cassette, sub-family A, member 4 ,biology.protein ,Hydroxyl radical ,ATP-Binding Cassette Transporters ,sense organs ,qAF, quantitative fundus autofluorescence - Abstract
The ability of iron to transfer electrons enables the contribution of this metal to a variety of cellular activities even as the redox properties of iron are also responsible for the generation of hydroxyl radicals (•OH), the most destructive of the reactive oxygen species. We previously showed that iron can promote the oxidation of bisretinoid by generating highly reactive hydroxyl radical (•OH). Now we report that preservation of iron regulation in the retina is not sufficient to prevent iron-induced bisretinoid oxidative degradation when blood iron levels are elevated in liver-specific hepcidin knockout mice. We obtained evidence for the perpetuation of Fenton reactions in the presence of the bisretinoid A2E and visible light. On the other hand, iron chelation by deferiprone was not associated with changes in postbleaching recovery of 11-cis-retinal or dark-adapted ERG b-wave amplitudes indicating that the activity of Rpe65, a rate-determining visual cycle protein that carries an iron-binding domain, is not affected. Notably, iron levels were elevated in the neural retina and retinal pigment epithelial (RPE) cells of Abca4−/− mice. Consistent with higher iron content, ferritin-L immunostaining was elevated in RPE of a patient diagnosed with ABCA4-associated disease and in RPE and photoreceptor cells of Abca4−/− mice. In neural retina of the mutant mice, reduced Tfrc mRNA was also an indicator of retinal iron overload. Thus iron chelation may defend retina when bisretinoid toxicity is implicated in disease processes.
- Published
- 2021
43. SYSTEMIC MEDICATION USE AND THE INCIDENCE AND GROWTH OF GEOGRAPHIC ATROPHY IN THE COMPARISON OF AGE-RELATED MACULAR DEGENERATION TREATMENTS TRIALS
- Author
-
Juan E. Grunwald, Brian L VanderBeek, Daniel F. Martin, Delu Song, Gui-Shuang Ying, Ebenezer Daniel, Maureen G. Maguire, Peiying Hua, and Joshua L. Dunaief
- Subjects
0301 basic medicine ,Male ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Time Factors ,genetic structures ,Bevacizumab ,Fundus Oculi ,Visual Acuity ,Angiogenesis Inhibitors ,Article ,03 medical and health sciences ,Macular Degeneration ,0302 clinical medicine ,Internal medicine ,Geographic Atrophy ,Ranibizumab ,medicine ,Humans ,Fluorescein Angiography ,Aged ,Aged, 80 and over ,Proportional hazards model ,business.industry ,Incidence (epidemiology) ,Incidence ,Hazard ratio ,General Medicine ,Macular degeneration ,medicine.disease ,eye diseases ,United States ,Clinical trial ,Ophthalmology ,030104 developmental biology ,Choroidal neovascularization ,Treatment Outcome ,Intravitreal Injections ,030221 ophthalmology & optometry ,Female ,sense organs ,medicine.symptom ,business ,Tomography, Optical Coherence ,medicine.drug ,Follow-Up Studies - Abstract
PURPOSE: To determine associations of systemic medications with the incidence and growth of geographic atrophy (GA) in participants of the comparison of age-related macular degeneration treatments trials. METHODS: Participants of comparison of age-related macular degeneration treatments trials with new untreated choroidal neovascularization in the study eye (one study eye per participant) were randomized to receive treatment with bevacizumab or ranibizumab. Participants were released from clinical trial treatment at 2 years and examined at approximately 5 years. Color fundus photographs and fluorescein angiograms taken at baseline, Years 1, 2, and 5 were assessed for the presence and size of GA by two masked graders. Participants were interviewed about systemic medication use at baseline. Systemic medications previously reported to be associated with age-related macular degeneration were evaluated for associations with GA incidence in study eye using univariable and multivariable Cox models and for association with the GA growth using linear mixed effects models. RESULTS: In multivariable analysis of 1,011 study eyes without baseline GA, systemic medications, including cholinesterase inhibitors, angiotensin-converting enzyme inhibitors, calcium channel blockers, beta-blockers, diuretics, aspirin, steroids, statins, hormone replacement therapy, antacids, and drugs targeting G protein-coupled receptors, were not associated with GA incidence in the study eye (all adjusted hazard ratios ≤1.86, P ≥ 0.18). In multivariable analysis of 214 study eyes with longitudinal GA size measurements, calcium channel blockers were associated with a higher GA growth rate (0.40 vs. 0.30 mm/year, P =0.02). CONCLUSION: None of the systemic medications analyzed were associated with GA incidence. However, calcium channel blockers were associated with a higher growth rate of GA in the study eye.
- Published
- 2020
44. GLP-1R agonist NLY01 reduces retinal inflammation, astrocyte reactivity, and retinal ganglion cell death secondary to ocular hypertension
- Author
-
Ahmara G. Ross, Katherine E. Uyhazi, Samyuktha Guttha, Albert R. Bargoud, Modupe O Adetunji, Qi N. Cui, Joshua L. Dunaief, and Jacob Sterling
- Subjects
Retina ,genetic structures ,Microglia ,business.industry ,Neurodegeneration ,Glaucoma ,medicine.disease ,Retinal ganglion ,eye diseases ,medicine.anatomical_structure ,Retinal ganglion cell ,medicine ,Cancer research ,sense organs ,business ,Neuroinflammation ,Astrocyte - Abstract
SUMMARYGlaucoma is the leading cause of irreversible blindness worldwide and is characterized by the death of retinal ganglion cells. Reduction of intraocular pressure (IOP) is the only therapeutic mechanism available to slow disease progression. However, glaucoma can continue to progress despite normalization of IOP. New treatments are needed to reduce vision loss and improve outcomes for patients who have exhausted existing therapeutic avenues. Recent studies have implicated neuroinflammation in the pathogenesis of neurodegenerative diseases of both the retina and the brain, including glaucoma and Parkinson’s disease. Pro-inflammatory A1 astrocytes contribute to neuronal cell death in multiple disease processes and have been targeted therapeutically in mouse models of Parkinson’s disease. Microglial release of pro-inflammatory cytokines C1q, IL-1α, and TNF-α is sufficient to drive the formation of A1 astrocytes. The role of A1 astrocytes in glaucoma pathogenesis has not been explored. Using a mouse model of glaucoma, we demonstrated that IOP elevation was sufficient to trigger production of C1q, IL-1α, and TNF-α by infiltrating macrophages followed by resident microglia. These three cytokines drove the formation of A1 astrocytes in the retina. Furthermore, cytokine production and A1 astrocyte transformation persisted following IOP normalization. Ablation of this pathway, by either genetic deletions of C1q, IL-1α, and TNF-α, or treatment with glucagon-like peptide-1 receptor agonist NLY01, reduced A1 astrocyte transformation and RGC death. Together, these results highlight a new neuroinflammatory mechanism behind glaucomatous neurodegeneration that can be therapeutically targeted by NLY01 administration.
- Published
- 2020
45. GLP-1 Receptor Agonist NLY01 Reduces Retinal Inflammation and Neuron Death Secondary to Ocular Hypertension
- Author
-
Samyuktha Guttha, Ahmara G. Ross, Albert Bargoud, Joshua L. Dunaief, Katherine E. Uyhazi, Qi N. Cui, Jacob Sterling, and Modupe O Adetunji
- Subjects
0301 basic medicine ,Agonist ,Male ,Retinal Ganglion Cells ,medicine.drug_class ,A1 reactive astrocyte ,Retinal ganglion ,General Biochemistry, Genetics and Molecular Biology ,Glucagon-Like Peptide-1 Receptor ,Article ,neuroinflammation ,03 medical and health sciences ,0302 clinical medicine ,Interleukin-1alpha ,medicine ,Animals ,retinal ganglion cell ,lcsh:QH301-705.5 ,Neuroinflammation ,Intraocular Pressure ,Inflammation ,CD11b Antigen ,Microglia ,Cell Death ,business.industry ,Tumor Necrosis Factor-alpha ,Complement C1q ,Neurodegeneration ,medicine.disease ,NLY01 ,Astrogliosis ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,glaucoma ,Retinal ganglion cell ,lcsh:Biology (General) ,Astrocytes ,Cancer research ,Female ,Ocular Hypertension ,sense organs ,Neuron death ,business ,030217 neurology & neurosurgery ,Retinal Neurons - Abstract
SUMMARY Glaucoma is the leading cause of irreversible blindness and is characterized by the death of retinal ganglion cells (RGCs). Recent studies have implicated pro-inflammatory microglia, macrophages, and A1 astrocytes in the pathogenesis of neurodegenerative diseases. The role of pro-inflammatory, neurotoxic A1 astrocytes in glaucoma is just beginning to be explored. Using a mouse model of glaucoma, we demonstrate that ocular hypertension is sufficient to trigger production of C1q, interleukin-1α (IL-1α), and tumor necrosis factor α (TNF-α), three cytokines necessary and sufficient to drive the formation of A1 astrocytes. Upregulation of these cytokines occurs first in CD11b+ CD11c+ cells followed by CD11b+ CD11c− cells. Ablation of this pathway, by either genetic deletions of C1qa, IL-1α, and TNF-α, or treatment with glucagon-like peptide-1 receptor agonist NLY01, reduces A1 astrocyte transformation and RGC death. Together, these results highlight a neuroinflammatory mechanism of glaucomatous neurodegeneration that can be therapeutically targeted by NLY01 administration., Graphical Abstract, In Brief Sterling et al. show that in response to ocular hypertension, CD11b+ CD11c+ cells contribute to early retinal inflammation and astrogliosis in a mouse model of glaucoma. Ablation of this inflammatory pathway, via genetic deletion or administration of the GLP-1R agonist NLY01, prevents astrogliosis and retinal ganglion cell death.
- Published
- 2020
46. Oral administration of the iron chelator deferiprone protects against loss of retinal ganglion cells in a mouse model of glaucoma
- Author
-
Albert Bargoud, Qi N. Cui, Ahmara G. Ross, Ying Song, and Joshua L. Dunaief
- Subjects
Male ,Retinal Ganglion Cells ,medicine.medical_specialty ,genetic structures ,Glaucoma ,Ocular hypertension ,Administration, Oral ,medicine.disease_cause ,Iron Chelating Agents ,Retinal ganglion ,Neuroprotection ,Article ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Ophthalmology ,Medicine ,Animals ,Deferiprone ,business.industry ,Retinal Degeneration ,Retinal ,Optic Nerve ,medicine.disease ,Sensory Systems ,eye diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,chemistry ,Optic nerve ,Female ,sense organs ,business ,Oxidative stress - Abstract
Glaucoma is a progressive neurodegenerative process affecting the retinal ganglion cells (RGCs) and the optic nerve. Oxidative stress has been implicated in glaucoma pathogenesis, and iron is a potent generator of oxidative stress. The oral iron chelator deferiprone (DFP) is protective against retinal degenerations associated with oxidative stress. To test whether DFP could be protective in glaucoma, we used microbead injections to induce elevated intraocular pressure (IOP) in a cohort of 3-month old C57BL/6J mice. One eye of each animal was injected with magnetic microbeads resulting in ocular hypertension for >7 weeks while the fellow eye was injected with saline and served as a normotensive internal control. While half of the cohort received oral DFP (1 mg/ml in the drinking water), the other half did not and served as controls. After 8 weeks, Brn3a immunolabeling of flat-mounted retinas was used for manual RGC quantification. Axon counts were obtained from thin sections of optic nerves using the AxonJ plugin for ImageJ. DFP administration was protective against RGC and optic nerve loss in the setting of elevated IOP. These results suggest that iron chelation by DFP may provide glaucoma neuroprotection.
- Published
- 2020
47. Interleukin-6 triggers toxic neuronal iron sequestration in response to pathological α-synuclein
- Author
-
Jacob K. Sterling, Tae-In Kam, Samyuktha Guttha, Hyejin Park, Bailey Baumann, Amir A. Mehrabani-Tabari, Hannah Schultz, Brandon Anderson, Ahab Alnemri, Shih-Ching Chou, Juan C. Troncoso, Valina L. Dawson, Ted M. Dawson, and Joshua L. Dunaief
- Subjects
Male ,Neurons ,Behavior, Animal ,Interleukin-6 ,Dopaminergic Neurons ,Iron ,Mice, Transgenic ,Parkinson Disease ,Iron Chelating Agents ,General Biochemistry, Genetics and Molecular Biology ,Mice, Inbred C57BL ,Substantia Nigra ,Disease Models, Animal ,Nerve Degeneration ,alpha-Synuclein ,Animals ,Female ,Signal Transduction - Abstract
α-synuclein (α-syn) aggregation and accumulation drive neurodegeneration in Parkinson's disease (PD). The substantia nigra of patients with PD contains excess iron, yet the underlying mechanism accounting for this iron accumulation is unclear. Here, we show that misfolded α-syn activates microglia, which release interleukin 6 (IL-6). IL-6, via its trans-signaling pathway, induces changes in the neuronal iron transcriptome that promote ferrous iron uptake and decrease cellular iron export via a pathway we term the cellular iron sequestration response, or CISR. The brains of patients with PD exhibit molecular signatures of the IL-6-mediated CISR. Genetic deletion of IL-6, or treatment with the iron chelator deferiprone, reduces pathological α-syn toxicity in a mouse model of sporadic PD. These data suggest that IL-6-induced CISR leads to toxic neuronal iron accumulation, contributing to synuclein-induced neurodegeneration.
- Published
- 2022
48. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions
- Author
-
John D. Lambris, Yafeng Li, Joshua L. Dunaief, Benjamin J. Kim, and Dimitrios C. Mastellos
- Subjects
0301 basic medicine ,genetic structures ,Legal blindness ,Retina ,Article ,Complement components ,Unmet needs ,Macular Degeneration ,03 medical and health sciences ,0302 clinical medicine ,Geographic Atrophy ,Humans ,Medicine ,Complement Activation ,business.industry ,Complement C5 ,Inflammasome ,Complement C3 ,Macular degeneration ,medicine.disease ,eye diseases ,Sensory Systems ,Complement system ,Complement (complexity) ,Geographic atrophy ,Ophthalmology ,030104 developmental biology ,030221 ophthalmology & optometry ,sense organs ,business ,Neuroscience ,medicine.drug - Abstract
Age-related macular degeneration (AMD) remains a major cause of legal blindness, and treatment for the geographic atrophy form of AMD is a significant unmet need. Dysregulation of the complement cascade is thought to be instrumental for AMD pathophysiology. In particular, C3 and C5 are pivotal components of the complement cascade and have become leading therapeutic targets for AMD. In this article, we discuss C3 and C5 in detail, including their roles in AMD, biochemical and structural aspects, locations of expression, and the functions of C3 and C5 fragments. Further, the article critically reviews developing therapeutics aimed at C3 and C5, underscoring the potential effects of broad inhibition of complement at the level of C3 versus more specific inhibition at C5. The relationships of complement biology to the inflammasome and microglia/macrophage activity are highlighted. Concepts of C3 and C5 biology will be emphasized, while we point out questions that need to be settled and directions for future investigations.
- Published
- 2021
49. Optical coherence tomography identifies outer retina thinning in frontotemporal degeneration
- Author
-
Murray Grossman, Benjamin J. Kim, Aaishah R. Raquib, Tomas S. Aleman, Ebenezer Daniel, Delu Song, Gui-Shuang Ying, Wei Pan, David J. Irwin, Joshua L. Dunaief, and Jennifer D. Leveque
- Subjects
Adult ,Male ,medicine.medical_specialty ,genetic structures ,Retina ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Atrophy ,Optical coherence tomography ,Ophthalmology ,medicine ,Humans ,Visual Pathways ,Outer nuclear layer ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Retinal ,Middle Aged ,medicine.disease ,eye diseases ,DNA-Binding Proteins ,Cross-Sectional Studies ,medicine.anatomical_structure ,ROC Curve ,chemistry ,Frontotemporal Dementia ,Disease Progression ,030221 ophthalmology & optometry ,Biomarker (medicine) ,Female ,sense organs ,Neurology (clinical) ,Tauopathy ,Frontotemporal Lobar Degeneration ,Mental Status Schedule ,business ,Tomography, Optical Coherence ,030217 neurology & neurosurgery ,Frontotemporal dementia - Abstract
Objective:Whereas Alzheimer disease (AD) is associated with inner retina thinning visualized by spectral-domain optical coherence tomography (SD-OCT), we sought to determine if the retina has a distinguishing biomarker for frontotemporal degeneration (FTD).Methods:Using a cross-sectional design, we examined retinal structure in 38 consecutively enrolled patients with FTD and 44 controls using a standard SD-OCT protocol. Retinal layers were segmented with the Iowa Reference Algorithm. Subgroups of highly predictive molecular pathology (tauopathy, TAR DNA–binding protein 43, unknown) were determined by clinical criteria, genetic markers, and a CSF biomarker (total tau: β-amyloid) to exclude presumed AD. We excluded eyes with poor image quality or confounding diseases. SD-OCT measures of patients (n = 46 eyes) and controls (n = 69 eyes) were compared using a generalized linear model accounting for intereye correlation, and correlations between retinal layer thicknesses and Mini-Mental State Examination (MMSE) were evaluated.Results:Adjusting for age, sex, and race, patients with FTD had a thinner outer retina than controls (132 vs 142 μm, p = 0.004). Patients with FTD also had a thinner outer nuclear layer (ONL) (88.5 vs 97.9 μm, p = 0.003) and ellipsoid zone (EZ) (14.5 vs 15.1 μm, p = 0.009) than controls, but had similar thicknesses for inner retinal layers. The outer retina thickness of patients correlated with MMSE (Spearman r = 0.44, p = 0.03). The highly predictive tauopathy subgroup (n = 31 eyes) also had a thinner ONL (88.7 vs 97.4 μm, p = 0.01) and EZ (14.4 vs 15.1 μm, p = 0.01) than controls.Conclusions:FTD is associated with outer retina thinning, and this thinning correlates with disease severity.
- Published
- 2017
50. Murine systemic thrombophilia and hemolytic uremic syndrome from a factor H point mutation
- Author
-
Damodar Gullipalli, Lin Zhou, Lawrence F. Brass, Delu Song, Imran Mohammed, Wen-Chao Song, Hong Wang, Sayaka Sato, Yoshiyasu Ueda, Joshua L. Dunaief, Takashi Miwa, X. Long Zheng, Yingying Mao, Yuan Wang, Zhongjian Cheng, Matthew Palmer, Shuchi Gupta, and Jialing Bao
- Subjects
0301 basic medicine ,Thrombotic microangiopathy ,Blotting, Western ,Immunology ,Thrombophilia ,Biochemistry ,Mice ,03 medical and health sciences ,Atypical hemolytic uremic syndrome ,medicine ,Humans ,Immunologic Factors ,Animals ,Point Mutation ,Complement Activation ,Mice, Knockout ,biology ,CD46 ,Thrombosis ,Cell Biology ,Hematology ,Platelets and Thrombopoiesis ,medicine.disease ,Complement system ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Interaction with host ,Complement Factor H ,Factor H ,Hemolytic-Uremic Syndrome ,biology.protein ,Factor D - Abstract
Complement plays a key role in host defense, but its dysregulation can cause autologous tissue injury. Complement activation is normally controlled by regulatory proteins, including factor H (FH) in plasma and membrane cofactor protein (MCP) on the cell surface. Mutations in FH and MCP are linked to atypical hemolytic uremic syndrome, a type of thrombotic microangiopathy (TMA) that causes renal failure. We describe here that disruption of FH function on the cell surface can also lead to disseminated complement-dependent macrovascular thrombosis. By gene targeting, we introduced a point mutation (W1206R) into murine FH that impaired its interaction with host cells but did not affect its plasma complement-regulating activity. Homozygous mutant mice carrying this mutation developed renal TMA as well as systemic thrombophilia involving large blood vessels in multiple organs, including liver, lung, spleen, and kidney. Approximately 30% of mutant mice displayed symptoms of stroke and ischemic retinopathy, and 48% died prematurely. Genetic deficiency of complement C3 and factor D prevented both the systemic thrombophilia and renal TMA phenotypes. These results demonstrate a causal relationship between complement dysregulation and systemic angiopathy and suggest that complement activation may contribute to various human thrombotic disorders involving both the micro- and macrovasculature.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.