Network


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

Hotspot


Dive into the research topics where Dong-Gyu Jo is active.

Publication


Featured researches published by Dong-Gyu Jo.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Calorie restriction up-regulates the plasma membrane redox system in brain cells and suppresses oxidative stress during aging

Dong-Hoon Hyun; Scott S. Emerson; Dong-Gyu Jo; Mark P. Mattson; Rafael de Cabo

The plasma membrane (PM) contains redox enzymes that provide electrons for energy metabolism and recycling of antioxidants such as coenzyme Q and α-tocopherol. Brain aging and neurodegenerative disorders involve impaired energy metabolism and oxidative damage, but the involvement of the PM redox system (PMRS) in these processes is unknown. Caloric restriction (CR), a manipulation that protects the brain against aging and disease, increased activities of PMRS enzymes (NADH-ascorbate free radical reductase, NADH-quinone oxidoreductase 1, NADH-ferrocyanide reductase, NADH-coenzyme Q10 reductase, and NADH-cytochrome c reductase) and antioxidant levels (α-tocopherol and coenzyme Q10) in brain PM during aging. Age-related increases in PM lipid peroxidation, protein carbonyls, and nitrotyrosine were attenuated by CR, levels of PMRS enzyme activities were higher, and markers of oxidative stress were lower in cultured neuronal cells treated with CR serum compared with those treated with ad libitum serum. These findings suggest important roles for the PMRS in protecting brain cells against age-related increases in oxidative and metabolic stress.


Cell Death and Disease | 2013

Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke

Yang-Wei Fann; S-Y Lee; Silvia Manzanero; Sung-Chun Tang; Mathias Gelderblom; Prasad Chunduri; Christian Bernreuther; Markus Glatzel; Yi-Lin Cheng; John Thundyil; Alexander Widiapradja; Ker Zhing Lok; S L Foo; Y-C Wang Wang; Y-I Li; Grant R. Drummond; Milan Basta; Tim Magnus; Dong-Gyu Jo; Mark P. Mattson; Christopher G. Sobey; Thiruma V. Arumugam

Multi-protein complexes called inflammasomes have recently been identified and shown to contribute to cell death in tissue injury. Intravenous immunoglobulin (IVIg) is an FDA-approved therapeutic modality used for various inflammatory diseases. The objective of this study is to investigate dynamic responses of the NLRP1 and NLRP3 inflammasomes in stroke and to determine whether the NLRP1 and NLRP3 inflammasomes can be targeted with IVIg for therapeutic intervention. Primary cortical neurons were subjected to glucose deprivation (GD), oxygen–glucose deprivation (OGD) or simulated ischemia-reperfusion (I/R). Ischemic stroke was induced in C57BL/6J mice by middle cerebral artery occlusion, followed by reperfusion. Neurological assessment was performed, brain tissue damage was quantified, and NLRP1 and NLRP3 inflammasome protein levels were evaluated. NLRP1 and NLRP3 inflammasome components were also analyzed in postmortem brain tissue samples from stroke patients. Ischemia-like conditions increased the levels of NLRP1 and NLRP3 inflammasome proteins, and IL-1β and IL-18, in primary cortical neurons. Similarly, levels of NLRP1 and NLRP3 inflammasome proteins, IL-1β and IL-18 were elevated in ipsilateral brain tissues of cerebral I/R mice and stroke patients. Caspase-1 inhibitor treatment protected cultured cortical neurons and brain cells in vivo in experimental stroke models. IVIg treatment protected neurons in experimental stroke models by a mechanism involving suppression of NLRP1 and NLRP3 inflammasome activity. Our findings provide evidence that the NLRP1 and NLRP3 inflammasomes have a major role in neuronal cell death and behavioral deficits in stroke. We also identified NLRP1 and NLRP3 inflammasome inhibition as a novel mechanism by which IVIg can protect brain cells against ischemic damage, suggesting a potential clinical benefit of therapeutic interventions that target inflammasome assembly and activity.


Biochemical and Biophysical Research Communications | 2009

Alzheimer’s disease and Notch signaling

Ha-Na Woo; Jong-Sung Park; A-Ryeong Gwon; Thiruma V. Arumugam; Dong-Gyu Jo

Cleavage of the amyloid precursor protein (APP) by gamma-secretase generates a neurotoxic amyloid beta-peptide (Abeta) that is thought to be associated with the neurodegeneration observed in Alzheimers disease (AD) patients. Presenilin is the catalytic member of the gamma-secretase proteolytic complex and mutations in presenilins are the major cause of early-onset familial Alzheimers disease. In addition to APP, gamma-secretase substrates include Notch1 homologues, Notch ligands Delta and Jagged, and additional type I membrane proteins, raising concerns about mechanism-based toxicities that might arise as a consequence of inhibiting gamma-secretase. Notch signaling is involved in tumorigenesis as well as in determining the fates of neural and nonneural cells during development and in adults. Alterations in proteolysis of the Notch by gamma-secretase could be involved in the pathogenesis of AD. Inconsistently, several recent observations have indicated that enhanced Notch signaling and expression could be instrumental in neurodegeneration in AD. Therefore, detailed and precise study of Notch signaling in AD is important for elucidating diverse mechanisms of pathogenesis and potentially for treating and preventing Alzheimers disease.


Stroke | 2011

Notch Activation Enhances the Microglia-Mediated Inflammatory Response Associated With Focal Cerebral Ischemia

Zelan Wei; Srinivasulu Chigurupati; Thiruma V. Arumugam; Dong-Gyu Jo; He Li; Sic L. Chan

Background and Purpose— Activation of Notch worsens ischemic brain damage as antisense knockdown or pharmacological inhibition of the Notch pathway reduces the infarct size and improves the functional outcome in a mouse model of stroke. We sought to determine whether Notch activation contributes to postischemic inflammation by directly modulating the microglial innate response. Methods— The microglial response and the attendant inflammatory reaction were evaluated in Notch1 antisense transgenic (Tg) and in nontransgenic (non-Tg) mice subjected to middle cerebral artery occlusion with or without treatment with a &ggr;-secretase inhibitor (GSI). To investigate the impact of Notch on microglial effector functions, primary mouse microglia and murine BV-2 microglial cell line were exposed to oxygen glucose deprivation or lipopolysaccharide in the presence or absence of GSI. Immunofluorescence labeling, Western blotting, and reverse-transcription polymerase chain reaction were performed to measure microglial activation and production of inflammatory cytokines. The nuclear translocation of nuclear factor-&kgr;B in microglia was assessed by immunohistochemistry. The neurotoxic potential of microglia was determined in cocultures. Results— Notch1 antisense mice exhibit significantly lower numbers of activated microglia and reduced proinflammatory cytokine expression in the ipsilateral ischemic cortices compared to non-Tg mice. Microglial activation also was attenuated in Notch1 antisense cultures and in non-Tg cultures treated with GSI. GSI significantly reduced nuclear factor-&kgr;B activation and expression of proinflammatory mediators and markedly attenuated the neurotoxic activity of microglia in cocultures. Conclusions— These findings establish a role for Notch signaling in modulating the microglia innate response and suggest that inhibition of Notch might represent a complementary therapeutic approach to prevent reactive gliosis in stroke and neuroinflammation-related degenerative disorders.


The FASEB Journal | 2001

Pro-apoptotic function of calsenilin/DREAM/KChIP3

Dong-Gyu Jo; Min Jung Kim; Yun Hee Choi; In-Ki Kim; Yu-Hyun Song; Ha-Na Woo; Chul-Woong Chung; Yong-Keun Jung

Apoptotic cell death and increased production of amyloid β peptide (Aβ) are pathological features of Alzheimer’s disease (AD), although the exact contribution of apoptosis to the pathogenesis of the disease remains unclear. Here we describe a novel pro‐apoptotic function of calsenilin/DREAM/KChIP3. By antisense oligonucleotide‐induced inhibition of calsenilin/DREAM/KChIP3 synthesis, apoptosis induced by Fas, Ca2+‐ionophore, or thapsigargin is attenuated. Conversely, calsenilin/DREAM/KChIP3 expression induced the morphological and biochemical features of apoptosis, including cell shrinkage, DNA laddering, and caspase activation. Calsenilin/DREAM/KChIP3‐induced apoptosis was suppressed by caspase inhibitor z‐VAD and by Bcl‐xL, and was potentiated by increasing cytosolic Ca2+, expression of Swedish amyloid precursor protein mutant (APPsw) or presenilin 2 (PS2), but not by a PS2 deletion lacking its C‐terminus (PS2/411stop). In addition, calsenilin/DREAM/KChIP3 expression increased Aβ42 production in cells expressing APPsw, which was potentiated by PS2, but not by PS2/411stop, which suggests a role for apoptosis‐associated Aβ42 production of calsenilin/DREAM/KChIP3.


Neurobiology of Aging | 2010

Evidence that γ-secretase mediates oxidative stress-induced β-secretase expression in Alzheimer's disease

Dong-Gyu Jo; Thiruma V. Arumugam; Ha-Na Woo; Jong-Sung Park; Sung-Chun Tang; Mohamed R. Mughal; Dong-Hoon Hyun; Jun-Hyung Park; Yun-Hyung Choi; A-Ryeong Gwon; Simonetta Camandola; Aiwu Cheng; Huaibin Cai; Weihong Song; William R. Markesbery; Mark P. Mattson

Beta-secretase (BACE1), an enzyme responsible for the production of amyloid beta-peptide (Abeta), is increased by oxidative stress and is elevated in the brains of patients with sporadic Alzheimers disease (AD). Here, we show that oxidative stress fails to induce BACE1 expression in presenilin-1 (gamma-secretase)-deficient cells and in normal cells treated with gamma-secretase inhibitors. Oxidative stress-induced beta-secretase activity and sAPPbeta levels were suppressed by gamma-secretase inhibitors. Levels of gamma- and beta-secretase activities were greater in brain tissue samples from AD patients compared to non-demented control subjects, and the elevated BACE1 level in the brains of 3xTgAD mice was reduced by treatment with a gamma-secretase inhibitor. Our findings suggest that gamma-secretase mediates oxidative stress-induced expression of BACE1 resulting in excessive Abeta production in AD.


Journal of Neurochemistry | 2004

Induction of pro-apoptotic calsenilin/DREAM/KChIP3 in Alzheimer's disease and cultured neurons after amyloid-β exposure

Dong-Gyu Jo; Joo-Yong Lee; Yeon-Mi Hong; Sungmin Song; Inhee Mook-Jung; Jae-Young Koh; Yong-Keun Jung

Calsenilin/DREAM/KChIP3 was identified as a calcium‐binding protein that interacts with presenilins, serves as a transcription repressor, and binds to the A‐type potassium channel. In this study, we hypothesized that calsenilin might be involved in the neurodegeneration of Alzheimers disease and examined calsenilin expression in Alzheimers disease. Calsenilin levels were elevated in the cortex region of Alzheimers patient brains and in the neocortex and the hippocampus of Swedish mutant β‐amyloid precursor protein transgenic mice brains. Induction of calsenilin was also observed in the activated astroglia as well as in the neurons surrounding β‐amyloid (Aβ)‐ and Congo red‐positive plaques. Exposing cultured cortical and hippocampal neurons to Aβ42, an amyloid‐β peptide whose deposition in the brain is a characteristic of Alzheimers disease, induced both calsenilin protein and mRNA expression, and cell death. Moreover, blocking the calsenilin expression protected the neuronal cells from Aβ toxicity. These findings suggest that chronic up‐regulation of calsenilin may be a risk factor for developing Alzheimers disease, perhaps by facilitating calsenilin‐mediated neurodegeneration.


Molecular Pharmacology | 2011

Evidence that γ-Secretase-Mediated Notch Signaling Induces Neuronal Cell Death via the Nuclear Factor-κB-Bcl-2-Interacting Mediator of Cell Death Pathway in Ischemic Stroke

Thiruma V. Arumugam; Yi-Lin Cheng; Yuri Choi; Yun-Hyung Choi; Sunghee Yang; Young-Kwang Yun; Jong-Sung Park; Dong Kwon Yang; John Thundyil; Mathias Gelderblom; Vardan T. Karamyan; Sung-Chun Tang; Sic L. Chan; Tim Magnus; Christopher G. Sobey; Dong-Gyu Jo

Notch-1 (Notch) is a cell surface receptor that regulates cell-fate decisions in the developing nervous system, and it may also have roles in synaptic plasticity in the adult brain. Binding of its ligands results in the proteolytic cleavage of Notch by the γ-secretase enzyme complex, thereby causing the release of a Notch intracellular domain (NICD) that translocates to the nucleus, in which it regulates transcription. Here we show that activation of Notch modulates ischemic neuronal cell death in vitro and in vivo. Specifically, our findings from the use of Notch-1 siRNA or the overexpression of NICD indicate that Notch activation contributes to cell death. Using modified NICD, we demonstrate an apoptosis-inducing function of NICD in both the nucleus and the cytosol. NICD transfection-induced cell death was reduced by blockade of calcium signaling, caspase activation, and Janus kinase signaling. Inhibition of the Notch-activating enzyme, γ-secretase, protected against ischemic neuronal cell death by targeting an apoptotic protease, cleaved caspase-3, nuclear factor-κB (NF-κB), and the pro-death BH3-only protein, Bcl-2-interacting mediator of cell death (Bim). Treatment of mice with a γ-secretase inhibitor, compound E, reduced infarct size and improved functional outcome in a model of focal ischemic stroke. Furthermore, γ-secretase inhibition reduced NICD, p-p65, and Bim levels in vivo. These findings suggest that Notch signaling endangers neurons after ischemic stroke by modulating the NF-κB, pro-death protein Bim, and caspase pathways.


Journal of Controlled Release | 2014

Polyplex-releasing microneedles for enhanced cutaneous delivery of DNA vaccine.

Nak Won Kim; Min Sang Lee; Kyu Ri Kim; Jung Eun Lee; Kyuri Lee; Jong Sung Park; Yoh Matsumoto; Dong-Gyu Jo; Haeshin Lee; Doo Sung Lee; Ji Hoon Jeong

Microneedle (MN)-based DNA vaccines have many advantages over conventional vaccines administered by hypodermic needles. However, an efficient strategy for delivering DNA vaccines to intradermal cells has not yet been established. Here, we report a new approach for delivering polyplex-based DNA vaccines using MN arrays coated with a pH-responsive polyelectrolyte multilayer assembly (PMA). This approach enabled rapid release of polyplex upon application to the skin. In addition to the polyplex-releasing MNs, we attempted to further maximize the vaccination by developing a polymeric carrier that targeted resident antigen presenting cells (APCs) rich in the intradermal area, as well as a DNA vaccine encoding a secretable fusion protein containing amyloid beta monomer (Aβ1-42), an antigenic determinant. The resulting vaccination system was able to successfully induce a robust humoral immune response compared to conventional subcutaneous injection with hypodermal needles. In addition, antigen challenge after immunization elicited an immediate and strong recall immune response due to immunogenic memory. These results suggest the potential utility of MN-based polyplex delivery systems for enhanced DNA vaccination.


Scientific Reports | 2015

Cancer Therapy Using Ultrahigh Hydrophobic Drug-Loaded Graphene Derivatives

Surajit Some; A-Ryeong Gwon; Eunhee Hwang; Gahee Bahn; Yeoheung Yoon; Young-Min Kim; Seol-Hee Kim; Sora Bak; Junghee Yang; Dong-Gyu Jo; Hyoyoung Lee

This study aimed to demonstrate that curcumin (Cur)-containing graphene composites have high anticancer activity. Specifically, graphene-derivatives were used as nanovectors for the delivery of the hydrophobic anticancer drug Cur based on pH dependence. Different Cur-graphene composites were prepared based on polar interactions between Cur and the number of oxygen-containing functional groups of respective starting materials. The degree of drug-loading was found to be increased by increasing the number of oxygen-containing functional groups in graphene-derivatives. We demonstrated a synergistic effect of Cur-graphene composites on cancer cell death (HCT 116) both in vitro and in vivo. As-prepared graphene quantum dot (GQD)-Cur composites contained the highest amount of Cur nano-particles and exhibited the best anticancer activity compared to the other composites including Cur alone at the same dose. This is the first example of synergistic chemotherapy using GQD-Cur composites simultaneous with superficial bioprobes for tumor imaging.

Collaboration


Dive into the Dong-Gyu Jo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thiruma V. Arumugam

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sang-Ha Baik

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Gahee Bahn

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Yong-Keun Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Yuri Choi

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Jihoon Han

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Youn Choi

Sungkyunkwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge