Network


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

Hotspot


Dive into the research topics where Keiko Kataoka is active.

Publication


Featured researches published by Keiko Kataoka.


Cell Death & Differentiation | 2014

Programmed necrosis, not apoptosis, is a key mediator of cell loss and DAMP-mediated inflammation in dsRNA-induced retinal degeneration

Yusuke Murakami; Hidetaka Matsumoto; Mi In Roh; Andrea Giani; Keiko Kataoka; Yuki Morizane; Maki Kayama; Aristomenis Thanos; Shunji Nakatake; Shoji Notomi; Toshio Hisatomi; Yasuhiro Ikeda; Tatsuro Ishibashi; Kip M. Connor; Joan W. Miller; Demetrios G. Vavvas

There is no known treatment for the dry form of an age-related macular degeneration (AMD). Cell death and inflammation are important biological processes thought to have central role in AMD. Here we show that receptor-interacting protein (RIP) kinase mediates necrosis and enhances inflammation in a mouse model of retinal degeneration induced by dsRNA, a component of drusen in AMD. In contrast to photoreceptor-induced apoptosis, subretinal injection of the dsRNA analog poly(Iu2009:u2009C) caused necrosis of the retinal pigment epithelium (RPE), as well as macrophage infiltration into the outer retinas. In Rip3−/− mice, both necrosis and inflammation were prevented, providing substantial protection against poly(Iu2009:u2009C)-induced retinal degeneration. Moreover, after poly(Iu2009:u2009C) injection, Rip3−/− mice displayed decreased levels of pro-inflammatory cytokines (such as TNF-α and IL-6) in the retina, and attenuated intravitreal release of high-mobility group box-1 (HMGB1), a major damage-associated molecular pattern (DAMP). In vitro, poly(Iu2009:u2009C)-induced necrosis were inhibited in Rip3-deficient RPE cells, which in turn suppressed HMGB1 release and dampened TNF-α and IL-6 induction evoked by necrotic supernatants. On the other hand, Rip3 deficiency did not modulate directly TNF-α and IL-6 production after poly(Iu2009:u2009C) stimulation in RPE cells or macrophages. Therefore, programmed necrosis is crucial in dsRNA-induced retinal degeneration and may promote inflammation by regulating the release of intracellular DAMPs, suggesting novel therapeutic targets for diseases such as AMD.


Investigative Ophthalmology & Visual Science | 2014

Strain Difference in Photoreceptor Cell Death After Retinal Detachment in Mice

Hidetaka Matsumoto; Keiko Kataoka; Pavlina Tsoka; Kip M. Connor; Joan W. Miller; Demetrios G. Vavvas

PURPOSEnTo evaluate the potential for mouse genetic background to effect photoreceptor cell death in response to experimental retinal detachment (RD).nnnMETHODSnRetinal detachment was induced in three inbred mouse strains (C57BL/6, BALB/c, and B6129SF2) by subretinal injection of sodium hyaluronate. A time course of photoreceptor cell death was assessed by TUNEL assay. Total photoreceptor cell death was analyzed through comparing the outer nuclear layer (ONL)/inner nuclear layer (INL) ratio 7 days post RD. Western blot analysis or quantitative real-time PCR (qPCR) were performed to assess cell death signaling, expression of endogenous neurotrophin, and levels of apoptosis inhibitors 24 hours after RD. Inflammatory cytokine secretion and inflammatory cell infiltration were quantified by ELISA and immunostaining, respectively.nnnRESULTSnThe peak of photoreceptor cell death after RD was at 24 hours in all strains. Photoreceptor cell death as well as monocyte chemoattractant protein 1 and interleukin 6 secretion at 24 hours after RD was the highest in BALB/c, followed in order of magnitude by C57BL/6 and B6129SF2. Conversely, nerve growth factor expression and ONL/INL ratio were the lowest in BALB/c. Apoptosis signaling was higher in C57BL/6, whereas necroptosis signaling was higher in C57BL/6 and BALB/c. Autophagic signaling was higher in BALB/c. X-linked inhibitor of apoptosis (XIAP) and survivin protein levels were lower in C57BL/6 and BALB/c, respectively. Macrophage/microglia infiltration was higher in C57BL/6 and BALB/c at 24 hours after RD.nnnCONCLUSIONSnPhotoreceptor cell death after RD was significantly different among the three strains, suggesting the presence of genetic factors that affect photoreceptor cell death after RD.


Cell Death and Disease | 2015

Macrophage- and RIP3-dependent inflammasome activation exacerbates retinal detachment-induced photoreceptor cell death

Keiko Kataoka; Hidetaka Matsumoto; Hiroki Kaneko; Shoji Notomi; Kimio Takeuchi; J H Sweigard; A Atik; Yusuke Murakami; Kip M. Connor; Hiroko Terasaki; Joan W. Miller; Demetrios G. Vavvas

Detachment of photoreceptors from the retinal pigment epithelium is seen in various retinal disorders, resulting in photoreceptor death and subsequent vision loss. Cell death results in the release of endogenous molecules that activate molecular platforms containing caspase-1, termed inflammasomes. Inflammasome activation in retinal diseases has been reported in some cases to be protective and in others to be detrimental, causing neuronal cell death. Moreover, the cellular source of inflammasomes in retinal disorders is not clear. Here, we demonstrate that patients with photoreceptor injury by retinal detachment (RD) have increased levels of cleaved IL-1β, an end product of inflammasome activation. In an animal model of RD, photoreceptor cell death led to activation of endogenous inflammasomes, and this activation was diminished by Rip3 deletion. The major source of Il1b expression was found to be infiltrating macrophages in the subretinal space, rather than dying photoreceptors. Inflammasome inhibition attenuated photoreceptor death after RD. Our data implicate the infiltrating macrophages as a source of damaging inflammasomes after photoreceptor detachment in a RIP3-dependent manner and suggest a novel therapeutic target for treatment of retinal diseases.


The FASEB Journal | 2014

The alternative complement pathway regulates pathological angiogenesis in the retina

J. Harry Sweigard; Ryoji Yanai; Philipp Gaissert; Magali Saint-Geniez; Keiko Kataoka; Aristomenis Thanos; Gregory L. Stahl; John D. Lambris; Kip M. Connor

A defining feature in proliferative retinopathies is the formation of pathological neovessels. In these diseases, the balance between neovessel formation and regression determines blindness, making the modulation of neovessel growth highly desirable. The role of the immune system in these retinopathies is of increasing interest, but it is not completely understood. We investigated the role of the alternative complement pathway during the formation and resolution of aberrant neovascularization. We used alternative complement pathway–deficient (Fb–/–) mice and age‐ and strain‐matched control mice to assess neovessel development and regression in an oxygen‐induced retinopathy (OIR) mouse model. In the control mice, we found increased transcription of Fb after OIR treatment. In the Fb–/– mice, we prepared retinal flatmounts and identified an increased number of neovessels, peaking at postnatal day 17 (P17; P=0.001). Subjecting human umbilical vein endothelial cells (HUVECs) to low oxygen, mimicking a characteristic of neovessels, decreased the expression of the complement inhibitor Cd55. Finally, using laser capture microdissection (LCM) to isolate the neovessels after OIR, we found decreased expression of Cd55 (P=0.005). Together, our data implicate the alternative complement pathway in facilitating neovessel clearance by down‐regulating the complement inhibitor Cd55 specifically on neovessels, allowing for their targeted removal while leaving the established vasculature intact.—Sweigard, J. H., Yanai, R., Gaissert, P., Saint‐Geniez, M., Kataoka, K., Thanos, A., Stahl, G. L., Lambris, J. D., Connor, K. M. The alternative complement pathway regulates pathological angiogenesis in the retina. FASEB J. 28, 3171–3182 (2014). www.fasebj.org


Cell Death and Disease | 2014

Mammalian STE20-like kinase 2, not kinase 1, mediates photoreceptor cell death during retinal detachment

Hidetaka Matsumoto; Yusuke Murakami; Keiko Kataoka; H Lin; Kip M. Connor; Joan W. Miller; Dawang Zhou; Joseph Avruch; Demetrios G. Vavvas

Photoreceptor cell death is the definitive cause of vision loss in retinal detachment (RD). Mammalian STE20-like kinase (MST) is a master regulator of both cell death and proliferation and a critical factor in development and tumorigenesis. However, to date the role of MST in neurodegeneration has not been fully explored. Utilizing MST1−/− and MST2−/− mice we identified MST2, but not MST1, as a regulator of photoreceptor cell death in a mouse model of RD. MST2−/− mice demonstrated significantly decreased photoreceptor cell death and outer nuclear layer (ONL) thinning after RD. Additionally, caspase-3 activation was attenuated in MST2−/− mice compared to control mice after RD. The transcription of p53 upregulated modulator of apoptosis (PUMA) and Fas was also reduced in MST2−/− mice post-RD. Retinas of MST2−/− mice displayed suppressed nuclear relocalization of phosphorylated YAP after RD. Consistent with the reduction of photoreceptor cell death, MST2−/− mice showed decreased levels of proinflammatory cytokines such as monocyte chemoattractant protein 1 and interleukin 6 as well as attenuated inflammatory CD11b cell infiltration during the early phase of RD. These results identify MST2, not MST1, as a critical regulator of caspase-mediated photoreceptor cell death in the detached retina and indicate its potential as a future neuroprotection target.


Science Translational Medicine | 2015

Inhibition of the alternative complement pathway preserves photoreceptors after retinal injury

J. Harry Sweigard; Hidetaka Matsumoto; Kaylee E. Smith; Leo A. Kim; Eleftherios I. Paschalis; Yoko Okonuki; Alexandra Castillejos; Keiko Kataoka; Eiichi Hasegawa; Ryoji Yanai; Deeba Husain; John D. Lambris; Demetrios G. Vavvas; Joan W. Miller; Kip M. Connor

The alternative complement pathway is activated in response to retinal injury, and inhibiting this pathway prevents complement-mediated photoreceptor cell death. Preventing photoreceptor death after retinal injury Retinal detachment and subsequent degeneration of the retina can lead to progressive visual decline due to death of photoreceptor cells, the major light-sensing cells within the eye. Early inflammatory mediators are up-regulated in the eye of patients with retinal detachment including components of the alternative complement pathway. Using a mouse model of retinal detachment, Sweigard et al. found that by blocking the alternative complement pathway through both genetic and pharmacological means, photoreceptors were protected from cell death. Degeneration of photoreceptors is a primary cause of vision loss worldwide, making the underlying mechanisms surrounding photoreceptor cell death critical to developing new treatment strategies. Retinal detachment, characterized by the separation of photoreceptors from the underlying retinal pigment epithelium, is a sight-threatening event that can happen in a number of retinal diseases. The detached photoreceptors undergo apoptosis and programmed necrosis. Given that photoreceptors are nondividing cells, their loss leads to irreversible visual impairment even after successful retinal reattachment surgery. To better understand the underlying disease mechanisms, we analyzed innate immune system regulators in the vitreous of human patients with retinal detachment and correlated the results with findings in a mouse model of retinal detachment. We identified the alternative complement pathway as promoting early photoreceptor cell death during retinal detachment. Photoreceptors down-regulate membrane-bound inhibitors of complement, allowing for selective targeting by the alternative complement pathway. When photoreceptors in the detached retina were removed from the primary source of oxygen and nutrients (choroidal vascular bed), the retina became hypoxic, leading to an up-regulation of complement factor B, a key mediator of the alternative pathway. Inhibition of the alternative complement pathway in knockout mice or through pharmacological means ameliorated photoreceptor cell death during retinal detachment. Our current study begins to outline the mechanism by which the alternative complement pathway facilitates photoreceptor cell death in the damaged retina.


PLOS ONE | 2016

AMPK-Activated Protein Kinase Suppresses Ccr2 Expression by Inhibiting the NF-κB Pathway in RAW264.7 Macrophages

Fumiaki Kumase; Kimio Takeuchi; Yuki Morizane; Jun Suzuki; Hidetaka Matsumoto; Keiko Kataoka; Ahmad Al-Moujahed; Daniel E. Maidana; Joan W. Miller; Demetrios G. Vavvas

C-C chemokine receptor 2 (Ccr2) is a key pro-inflammatory marker of classic (M1) macrophage activation. Although Ccr2 is known to be expressed both constitutively and inductively, the full regulatory mechanism of its expression remains unclear. AMP-activated protein kinase (AMPK) is not only a master regulator of energy homeostasis but also a central regulator of inflammation. In this study, we sought to assess AMPK’s role in regulating RAW264.7 macrophage Ccr2 protein levels in resting (M0) or LPS-induced M1 states. In both M0 and M1 RAW264.7 macrophages, knockdown of the AMPKα1 subunit by siRNA led to increased Ccr2 levels whereas pharmacologic (A769662) activation of AMPK, attenuated LPS-induced increases in Ccr2 expression in an AMPK dependent fashion. The increases in Ccr2 levels by AMPK downregulation were partially reversed by NF-κB inhibition whereas TNF-a inhibition had minimal effects. Our results indicate that AMPK is a negative regulator of Ccr2 expression in RAW264.7 macrophages, and that the mechanism of action of AMPK inhibition of Ccr2 is mediated, in part, through the NF-κB pathway.


Investigative Ophthalmology & Visual Science | 2015

A Novel ImageJ Macro for Automated Cell Death Quantitation in the Retina

Daniel E. Maidana; Pavlina Tsoka; Bo Tian; Bernard Dib; Hidetaka Matsumoto; Keiko Kataoka; Haijiang Lin; Joan W. Miller; Demetrios G. Vavvas

PURPOSEnTUNEL assay is widely used to evaluate cell death. Quantification of TUNEL-positive (TUNEL+) cells in tissue sections is usually performed manually, ideally by two masked observers. This process is time consuming, prone to measurement errors, and not entirely reproducible. In this paper, we describe an automated quantification approach to address these difficulties.nnnMETHODSnWe developed an ImageJ macro to quantitate cell death by TUNEL assay in retinal cross-section images. The script was coded using IJ1 programming language. To validate this tool, we selected a dataset of TUNEL assay digital images, calculated layer area and cell count manually (done by two observers), and compared measurements between observers and macro results.nnnRESULTSnThe automated macro segmented outer nuclear layer (ONL) and inner nuclear layer (INL) successfully. Automated TUNEL+ cell counts were in-between counts of inexperienced and experienced observers. The intraobserver coefficient of variation (COV) ranged from 13.09% to 25.20%. The COV between both observers was 51.11 ± 25.83% for the ONL and 56.07 ± 24.03% for the INL. Comparing observers results with macro results, COV was 23.37 ± 15.97% for the ONL and 23.44 ± 18.56% for the INL.nnnCONCLUSIONSnWe developed and validated an ImageJ macro that can be used as an accurate and precise quantitative tool for retina researchers to achieve repeatable, unbiased, fast, and accurate cell death quantitation. We believe that this standardized measurement tool could be advantageous to compare results across different research groups, as it is freely available as open source.


Cell Death and Disease | 2015

Membrane-bound and soluble Fas ligands have opposite functions in photoreceptor cell death following separation from the retinal pigment epithelium.

Hidetaka Matsumoto; Yusuke Murakami; Keiko Kataoka; Shoji Notomi; D. Mantopoulos; George Trichonas; Joan W. Miller; Meredith S. Gregory; Bruce R. Ksander; Ann Marshak-Rothstein; Demetrios G. Vavvas

Fas ligand (FasL) triggers apoptosis of Fas-positive cells, and previous reports described FasL-induced cell death of Fas-positive photoreceptors following a retinal detachment. However, as FasL exists in membrane-bound (mFasL) and soluble (sFasL) forms, and is expressed on resident microglia and infiltrating monocyte/macrophages, the current study examined the relative contribution of mFasL and sFasL to photoreceptor cell death after induction of experimental retinal detachment in wild-type, knockout (FasL−/−), and mFasL-only knock-in (ΔCS) mice. Retinal detachment in FasL−/− mice resulted in a significant reduction of photoreceptor cell death. In contrast, ΔCS mice displayed significantly more apoptotic photoreceptor cell death. Photoreceptor loss in ΔCS mice was inhibited by a subretinal injection of recombinant sFasL. Thus, Fas/FasL-triggered cell death accounts for a significant amount of photoreceptor cell loss following the retinal detachment. The function of FasL was dependent upon the form of FasL expressed: mFasL triggered photoreceptor cell death, whereas sFasL protected the retina, indicating that enzyme-mediated cleavage of FasL determines, in part, the extent of vision loss following the retinal detachment. Moreover, it also indicates that treatment with sFasL could significantly reduce photoreceptor cell loss in patients with retinal detachment.


Investigative Ophthalmology & Visual Science | 2016

NMDA-induced Retinal Excitotoxicity Triggers Inflammation and Inflammasome Activation in Mice.

Pavlina Tsoka; Keiko Kataoka; Joan W. Miller; Demetrios G. Vavvas

Collaboration


Dive into the Keiko Kataoka's collaboration.

Top Co-Authors

Avatar

Demetrios G. Vavvas

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Joan W. Miller

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kip M. Connor

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Pavlina Tsoka

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Kimio Takeuchi

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel E. Maidana

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Shoji Notomi

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge