Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Joshua L. Dunaief is active.

Publication


Featured researches published by Joshua L. Dunaief.


Cell | 1994

The retinoblastoma protein and BRG1 form a complex and cooperate to induce cell cycle arrest

Joshua L. Dunaief; Bruce E. Strober; Sushovan Guha; Paul A. Khavari; Kimona Ålin; Jeremy Luban; Martin Begemann; Gerald R. Crabtree; S P Goff

The retinoblastoma tumor suppressor protein (RB) binds several cellular proteins involved in cell cycle progression. Using the yeast two-hybrid system, we found that RB bound specifically to the protein BRG1. BRG1 shares extensive sequence similarity to Drosophila brahma, an activator of homeotic gene expression, and the yeast transcriptional activator SNF2/SW12. BRG1 contains an RB-binding motif found in viral oncoproteins and bound to the A/B pocket and the hypophosphorylated form of RB. BRG1 did not bind RB in viral oncoprotein-transformed cells. Coimmunoprecipitation experiments suggested BRG1 associates with the RB family in vivo. In the human carcinoma cell line SW13, BRG1 exhibited tumor suppressor activity by inducing formation of flat, growth-arrested cells. This activity depended on the ability of BRG1 to cooperate and complex with RB, as both an RB-nonbinding mutant of BRG1 and the sequestration of RB by adenovirus E1A protein abolished flat cell formation.


Nature | 2011

DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration

Hiroki Kaneko; Sami Dridi; Valeria Tarallo; Bradley D. Gelfand; Benjamin J. Fowler; Won Gil Cho; Mark E. Kleinman; Steven L. Ponicsan; William W. Hauswirth; Vince A. Chiodo; Katalin Karikó; Jae-Wook Yoo; Dong-ki Lee; Majda Hadziahmetovic; Ying Qing Song; Smita Misra; Gautam Chaudhuri; Frank W. Buaas; Robert E. Braun; David R. Hinton; Qing-qing Zhang; Hans E. Grossniklaus; Jan M. Provis; Michele C. Madigan; Ann H. Milam; Nikki L. Justice; Romulo Albuquerque; Alexander D. Blandford; Sasha Bogdanovich; Yoshio Hirano

Geographic atrophy (GA), an untreatable advanced form of age-related macular degeneration, results from retinal pigmented epithelium (RPE) cell degeneration. Here we show that the microRNA (miRNA)-processing enzyme DICER1 is reduced in the RPE of humans with GA, and that conditional ablation of Dicer1, but not seven other miRNA-processing enzymes, induces RPE degeneration in mice. DICER1 knockdown induces accumulation of Alu RNA in human RPE cells and Alu-like B1 and B2 RNAs in mouse RPE. Alu RNA is increased in the RPE of humans with GA, and this pathogenic RNA induces human RPE cytotoxicity and RPE degeneration in mice. Antisense oligonucleotides targeting Alu/B1/B2 RNAs prevent DICER1 depletion-induced RPE degeneration despite global miRNA downregulation. DICER1 degrades Alu RNA, and this digested Alu RNA cannot induce RPE degeneration in mice. These findings reveal a miRNA-independent cell survival function for DICER1 involving retrotransposon transcript degradation, show that Alu RNA can directly cause human pathology, and identify new targets for a major cause of blindness.


Cell | 2012

DICER1 Loss and Alu RNA Induce Age-Related Macular Degeneration via the NLRP3 Inflammasome and MyD88

Valeria Tarallo; Yoshio Hirano; Bradley D. Gelfand; Sami Dridi; Nagaraj Kerur; Younghee Kim; Won Gil Cho; Hiroki Kaneko; Benjamin J. Fowler; Sasha Bogdanovich; Romulo Albuquerque; William W. Hauswirth; Vince A. Chiodo; Jennifer F. Kugel; James A. Goodrich; Steven L. Ponicsan; Gautam Chaudhuri; Michael P. Murphy; Joshua L. Dunaief; Balamurali K. Ambati; Yuichiro Ogura; Jae Wook Yoo; Dong Ki Lee; Patrick Provost; David R. Hinton; Gabriel Núñez; Judit Z. Baffi; Mark E. Kleinman; Jayakrishna Ambati

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Molecular and Cellular Biology | 1996

Functional interactions between the hBRM/hBRG1 transcriptional activators and the pRB family of proteins.

Bruce E. Strober; Joshua L. Dunaief; Guha; Stephen P. Goff

hBRG1 and hBRM are mammalian homologs of the SNF2/SW12 yeast transcriptional activator. These proteins exist in a large multisubunit complex that likely serves to remodel chromatin and, in so doing, facilitates the function of specific transcription factors. The retinoblastoma protein (pRB) inhibits cell cycle progression by repressing transcription of specific growth-related genes. Using the yeast two-hybrid system, we demonstrate that the members of the hBRG1/hBRM family of proteins interact with the pRB family of proteins, which includes pRB, p107, and p130. Interaction between the hBRG1/hBRM family with the pRB family likely influences cellular proliferation, as both hBRG1 and hBRM, but not mutants of these proteins unable to bind to pRB family members, inhibit the formation of drug-resistant colonies when transfected into the SW13 human adenocarcinoma cell line, which lacks endogenous hBRG1 or hBRM. Further, hBRM and two isoforms of hBRG1 induce the formation of flat, growth-arrested cells in a pRB family-dependent manner when introduced into SW13 cells. This flat-cell inducing activity is severely reduced by cotransfection of the wild-type E1A protein and variably reduced by the cotransfection of mutants of E1A that lack the ability to bind to some or all members of the pRB family.


Photochemistry and Photobiology | 2004

Mitochondria‐derived Reactive Oxygen Species Mediate Blue Light‐induced Death of Retinal Pigment Epithelial Cells

Ayala King; Eyal Gottlieb; David G. Brooks; Michael P. Murphy; Joshua L. Dunaief

Throughout the lifetime of an individual, light is focused onto the retina. The resulting photooxidative stress can cause acute or chronic retinal damage. The pathogenesis of age‐related macular degeneration (AMD), the leading cause of legal blindness in the developed world, involves oxidative stress and death of the retinal pigment epithelium (RPE) followed by death of the overlying photoreceptors. Evidence suggests that damage due to exposure to light plays a role in AMD and other age‐related eye diseases. In this work a system for lightinduced damage and death of the RPE, based on the human ARPE‐19 cell line, was used. Induction of mitochondriaderived reactive oxygen species (ROS) is shown to play a critical role in the death of cells exposed to short‐wavelength blue light (425 ± 20 nm). ROS and cell death are blocked either by inhibiting the mitochondrial electron transport chain or by mitochondria‐specific antioxidants. These results show that mitochondria are an important source of toxic oxygen radicals in blue light‐exposed RPE cells and may indicate new approaches for treating AMD using mitochondria‐targeted antioxidants.


Progress in Retinal and Eye Research | 2007

Iron homeostasis and toxicity in retinal degeneration

Xining He; Paul Hahn; Jared Iacovelli; Robert Wong; Chih King; Robert B. Bhisitkul; Mina Massaro-Giordano; Joshua L. Dunaief

Iron is essential for many metabolic processes but can also cause damage. As a potent generator of hydroxyl radical, the most reactive of the free radicals, iron can cause considerable oxidative stress. Since iron is absorbed through diet but not excreted except through menstruation, total body iron levels buildup with age. Macular iron levels increase with age, in both men and women. This iron has the potential to contribute to retinal degeneration. Here we present an overview of the evidence suggesting that iron may contribute to retinal degenerations. Intraocular iron foreign bodies cause retinal degeneration. Retinal iron buildup resulting from hereditary iron homeostasis disorders aceruloplasminemia, Friedreichs ataxia, and panthothenate kinase-associated neurodegeneration cause retinal degeneration. Mice with targeted mutation of the iron exporter ceruloplasmin have age-dependent retinal iron overload and a resulting retinal degeneration with features of age-related macular degeneration (AMD). Post mortem retinas from patients with AMD have more iron and the iron carrier transferrin than age-matched controls. Over the past 10 years much has been learned about the intricate network of proteins involved in iron handling. Many of these, including transferrin, transferrin receptor, divalent metal transporter-1, ferritin, ferroportin, ceruloplasmin, hephaestin, iron-regulatory protein, and histocompatibility leukocyte antigen class I-like protein involved in iron homeostasis (HFE) have been found in the retina. Some of these proteins have been found in the cornea and lens as well. Levels of the iron carrier transferrin are high in the aqueous and vitreous humors. The functions of these proteins in other tissues, combined with studies on cultured ocular tissues, genetically engineered mice, and eye exams on patients with hereditary iron diseases provide clues regarding their ocular functions. Iron may play a role in a broad range of ocular diseases, including glaucoma, cataract, AMD, and conditions causing intraocular hemorrhage. While iron deficiency must be prevented, the therapeutic potential of limiting iron-induced ocular oxidative damage is high. Systemic, local, or topical iron chelation with an expanding repertoire of drugs has clinical potential.


Journal of Clinical Investigation | 2011

mTOR-mediated dedifferentiation of the retinal pigment epithelium initiates photoreceptor degeneration in mice

Chen Zhao; Douglas Yasumura; Xiyan Li; Michael T. Matthes; Marcia Lloyd; Gregory Nielsen; Kelly Ahern; Michael Snyder; Dean Bok; Joshua L. Dunaief; Matthew M. LaVail; Douglas Vollrath

Retinal pigment epithelial (RPE) cell dysfunction plays a central role in various retinal degenerative diseases, but knowledge is limited regarding the pathways responsible for adult RPE stress responses in vivo. RPE mitochondrial dysfunction has been implicated in the pathogenesis of several forms of retinal degeneration. Here we have shown that postnatal ablation of RPE mitochondrial oxidative phosphorylation in mice triggers gradual epithelium dedifferentiation, typified by reduction of RPE-characteristic proteins and cellular hypertrophy. The electrical response of the retina to light decreased and photoreceptors eventually degenerated. Abnormal RPE cell behavior was associated with increased glycolysis and activation of, and dependence upon, the hepatocyte growth factor/met proto-oncogene pathway. RPE dedifferentiation and hypertrophy arose through stimulation of the AKT/mammalian target of rapamycin (AKT/mTOR) pathway. Administration of an oxidant to wild-type mice also caused RPE dedifferentiation and mTOR activation. Importantly, treatment with the mTOR inhibitor rapamycin blunted key aspects of dedifferentiation and preserved photoreceptor function for both insults. These results reveal an in vivo response of the mature RPE to diverse stressors that prolongs RPE cell survival at the expense of epithelial attributes and photoreceptor function. Our findings provide a rationale for mTOR pathway inhibition as a therapeutic strategy for retinal degenerative diseases involving RPE stress.


Investigative Ophthalmology & Visual Science | 2008

Foveolar Choroidal Circulation and Choroidal Neovascularization in Age-Related Macular Degeneration

Tatyana I. Metelitsina; Juan E. Grunwald; Joan DuPont; Gui-shuang Ying; Alexander J. Brucker; Joshua L. Dunaief

PURPOSE To investigate in a longitudinal study whether foveolar choroidal blood flow changes are associated with the development of choroidal neovascularization (CNV) in AMD. METHODS Relative foveolar choroidal blood velocity (ChBVel), volume (ChBVol), and flow (ChBFlow) were assessed in 135 patients with AMD, at baseline and then annually with laser Doppler flowmetry. All study eyes had visual acuity of 20/40 or better and no CNV at the time of enrollment. Comparison of foveolar choroidal circulatory measurements at baseline and their change before the development of CNV was made between eyes that had CNV and those that did not. RESULTS CNV developed in 28 eyes during the study. Baseline average foveolar ChBVol and ChBFlow in these eyes were 24% (P < 0.0001) and 20% (P = 0.0007) lower than that observed in the 165 eyes in which CNV did not develop. In the eyes with CNV, foveolar ChBVol and ChBFlow decreased by 9.6% and 11.5% before the formation of CNV, whereas in the eyes that did not, they increased by 6.7% (P = 0.006) and 2.8% (P = 0.004), respectively. Eyes with lower baseline foveolar ChBFlow were more likely (risk ratio = 3.47, 95% CI: 1.24-8.70) to show visual loss of three or more lines than were eyes with a higher baseline ChBFlow (P = 0.005). CONCLUSIONS The development of CNV and visual loss are associated with lower choroidal circulatory parameters at baseline. In addition, the results suggest that decreases in the foveolar choroidal circulation precede the development of CNV in AMD and may play some role in its development.


Free Radical Biology and Medicine | 2008

Oxidative stress in diseases of the human cornea

Akiva Shoham; Majda Hadziahmetovic; Joshua L. Dunaief; Marc B. Mydlarski; Hyman M. Schipper

Intense exposure to light, robust metabolic activity, and high oxygen tension render the human eye particularly vulnerable to oxidative damage and the list of ophthalmological disorders implicating reactive oxygen and nitrogen species is rapidly expanding. Here, we review the roles of oxidative stress in the etiopathogeneses and pathophysiology of diseases of the human cornea including pterygium, keratoconus, trauma and chemical injury, and a host of inflammatory, metabolic, degenerative, and iatrogenic conditions. Data from animal and tissue culture experimentation germane to these conditions are also adduced.


Retina-the Journal of Retinal and Vitreous Diseases | 2007

Iron toxicity as a potential factor in AMD.

Robert W. Wong; D Chimene Richa; Paul Hahn; W. Richard Green; Joshua L. Dunaief

While it has been known for years that iron overload is associated with retinal degeneration in the context of ocular siderosis, intraocular hemorrhage, and the hereditary diseases aceruloplasminemia and pantothenate kinase associated neurodegeneration, recent evidence suggests that age-related macular degeneration (AMD) may also be exacerbated by retinal iron overload. In the retina, iron is necessary for normal cellular function. Iron overload, however, can cause retinal toxicity through the generation of oxygen free radicals. Histopathology of eyes with macular degeneration has shown elevated levels of iron in the retinal pigment epithelium, Bruch membrane, and within drusen, some of which was chelatable in vitro with deferoxamine. In this review, the authors summarize the evidence that iron overload may contribute to AMD pathogenesis. It is hoped that continued investigation of the role of iron and iron associated proteins in the retina will uncover clues to AMD pathogenesis and lead to new preventative or therapeutic options.

Collaboration


Dive into the Joshua L. Dunaief's collaboration.

Top Co-Authors

Avatar

Delu Song

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ying Song

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tzvete Dentchev

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Yafeng Li

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jared Iacovelli

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Natalie Wolkow

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Gui-shuang Ying

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wen-Chao Song

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge