22 results on '"Joon-Il Jun"'
Search Results
2. CCN1 is an opsonin for bacterial clearance and a direct activator of Toll-like receptor signaling
- Author
-
Joon-Il Jun and Lester F. Lau
- Subjects
Science - Abstract
CCN1 is a matricellular protein with a variety of functions, including an effect on wound healing and an association with inflammation. Here, the authors identify a possible mechanism by showing that CCN1 mediates the clearance of bacterial infections in mice and activates TLR signalling.
- Published
- 2020
- Full Text
- View/download PDF
3. CCN1 coordinately regulates intestinal stem cell proliferation and differentiation through integrins αvβ3/αvβ5
- Author
-
Jonghoon Won, Jacob Choi, and Joon-Il Jun
- Subjects
digestive system - Abstract
Intestinal stem cells (ISCs) at the crypt base contribute to intestinal homeostasis through a balance between self-renewal and differentiation. However, the molecular mechanisms regulating this homeostatic balance remain elusive. Here we show that the matricellular protein CCN1/CYR61 coordinately regulates ISC proliferation and differentiation through distinct pathways emanating from CCN1 interaction with integrins αvβ3/αvβ5. Mice that delete Ccn1 in Lgr5+ ISCs or express mutant CCN1 unable to bind integrins αvβ3/αvβ5 exhibited exuberant ISC expansion and enhanced differentiation into secretory cells at the expense of absorptive enterocytes in the small intestine, leading to nutrient malabsorption. Analysis of crypt organoids revealed that through integrins αvβ3/αvβ5, CCN1 induces NF-κB-dependent Jag1 expression to regulate Notch activation for differentiation and promotes Src-mediated YAP activation and Dkk1 expression to control Wnt signaling for proliferation. Moreover, CCN1 and YAP amplify the activities of each other in a regulatory loop. These findings establish CCN1 as a novel niche factor in the intestinal crypts, providing new insights into how matrix signaling exerts overarching control of ISC homeostasis.
- Published
- 2021
- Full Text
- View/download PDF
4. CCN1 interacts with integrins to regulate intestinal stem cell proliferation and differentiation
- Author
-
Jonghoon Won, Joon-Il Jun, and Jacob Choi
- Subjects
Intestines ,Integrins ,Mice ,Multidisciplinary ,General Physics and Astronomy ,Animals ,Cell Differentiation ,General Chemistry ,Intestinal Mucosa ,Wnt Signaling Pathway ,General Biochemistry, Genetics and Molecular Biology ,Cell Proliferation ,Cysteine-Rich Protein 61 - Abstract
Intestinal stem cells (ISCs) at the crypt base contribute to intestinal homeostasis through a balance between self-renewal and differentiation. However, the molecular mechanisms regulating this homeostatic balance remain elusive. Here we show that the matricellular protein CCN1/CYR61 coordinately regulates ISC proliferation and differentiation through distinct pathways emanating from CCN1 interaction with integrins αvβ3/αvβ5. Mice that delete Ccn1 in Lgr5 + ISCs or express mutant CCN1 unable to bind integrins αvβ3/αvβ5 exhibited exuberant ISC expansion and enhanced differentiation into secretory cells at the expense of absorptive enterocytes in the small intestine, leading to nutrient malabsorption. Analysis of crypt organoids revealed that through integrins αvβ3/αvβ5, CCN1 induces NF-κB-dependent Jag1 expression to regulate Notch activation for differentiation and promotes Src-mediated YAP activation and Dkk1 expression to control Wnt signaling for proliferation. Moreover, CCN1 and YAP amplify the activities of each other in a regulatory loop. These findings establish CCN1 as a niche factor in the intestinal crypts, providing insights into how matrix signaling exerts overarching control of ISC homeostasis.
- Published
- 2021
5. CCN1 is an opsonin for bacterial clearance and a direct activator of Toll-like receptor signaling
- Author
-
Lester F. Lau and Joon Il Jun
- Subjects
Male ,0301 basic medicine ,Molecular biology ,General Physics and Astronomy ,Mice ,0302 clinical medicine ,Sf9 Cells ,Gene Knock-In Techniques ,lcsh:Science ,Toll-like receptor ,Multidisciplinary ,Molecular medicine ,integumentary system ,Chemistry ,Toll-Like Receptors ,Matricellular protein ,Pattern recognition receptor ,Opsonin Proteins ,Staphylococcal Infections ,3. Good health ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,CYR61 ,Pseudomonas aeruginosa ,Female ,Disease Susceptibility ,medicine.symptom ,Signal Transduction ,Methicillin-Resistant Staphylococcus aureus ,Cell biology ,Science ,Immunology ,Mice, Transgenic ,Inflammation ,Microbiology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Phagocytosis ,medicine ,Animals ,Humans ,Pseudomonas Infections ,Opsonin ,Pathogen-Associated Molecular Pattern Molecules ,General Chemistry ,Integrin alphaVbeta3 ,Mice, Inbred C57BL ,Disease Models, Animal ,TLR2 ,030104 developmental biology ,TLR4 ,lcsh:Q ,Cysteine-Rich Protein 61 - Abstract
Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvβ3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvβ3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair., CCN1 is a matricellular protein with a variety of functions, including an effect on wound healing and an association with inflammation. Here, the authors identify a possible mechanism by showing that CCN1 mediates the clearance of bacterial infections in mice and activates TLR signalling.
- Published
- 2020
- Full Text
- View/download PDF
6. CCN2 induces cellular senescence in fibroblasts
- Author
-
Joon Il Jun and Lester F. Lau
- Subjects
0301 basic medicine ,Senescence ,integumentary system ,Cell Biology ,Matrix metalloproteinase ,Biology ,medicine.disease ,Biochemistry ,Cell biology ,CTGF ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Downregulation and upregulation ,Fibrosis ,030220 oncology & carcinogenesis ,medicine ,Wound healing ,Fibroblast ,Molecular Biology ,Myofibroblast ,Research Article - Abstract
The expression of Ccn2 (CTGF) has been linked to fibrosis in many tissues and pathologies, although its activities in fibroblastic cells and precise mechanism of action in fibrogenesis are still controversial. Here, we showed that CCN2 can induce cellular senescence in fibroblasts both in vitro and in vivo, whereupon senescent cells express an anti-fibrotic “senescence-associated secretory phenotype” (SASP) that includes upregulation of matrix metalloproteinases and downregulation of collagen. Mechanistically, CCN2 induces fibroblast senescence through integrin α6β1-mediated accumulation of reactive oxygen species, leading to activation of p53 and induction of p16INK4a. In cutaneous wound healing, Ccn2 expression is highly elevated only during the initial inflammatory phase and quickly declines thereafter to a low level during the proliferation and maturation phases of healing when myofibroblasts play a major role. Consistent with this expression kinetics, knockdown of Ccn2 has little effect on the rate of wound closure, formation of senescent cells, or collagen content of the wounds. However, application of purified CCN2 protein on cutaneous wounds leads to induction of senescent cells, expression of SASP, and reduction of collagen content. These results show that CCN2 can induce cellular senescence in fibroblasts and is capable of exerting an anti-fibrotic effect in a context-dependent manner.
- Published
- 2016
- Full Text
- View/download PDF
7. Resolution of organ fibrosis
- Author
-
Lester F. Lau and Joon Il Jun
- Subjects
0301 basic medicine ,Senescence ,Apoptosis ,Matrix (biology) ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,Humans ,Myofibroblasts ,Loss function ,Cellular Senescence ,business.industry ,Review Series ,General Medicine ,Cell Dedifferentiation ,medicine.disease ,Cellular Reprogramming ,Cell biology ,Extracellular Matrix ,030104 developmental biology ,030220 oncology & carcinogenesis ,Wound healing ,business ,Myofibroblast ,Reprogramming - Abstract
Fibrosis is the excessive accumulation of extracellular matrix that often occurs as a wound healing response to repeated or chronic tissue injury, and may lead to the disruption of organ architecture and loss of function. Although fibrosis was previously thought to be irreversible, recent evidence indicates that certain circumstances permit the resolution of fibrosis when the underlying causes of injury are eradicated. The mechanism of fibrosis resolution encompasses degradation of the fibrotic extracellular matrix as well as elimination of fibrogenic myofibroblasts through their adaptation of various cell fates, including apoptosis, senescence, dedifferentiation, and reprogramming. In this Review, we discuss the present knowledge and gaps in our understanding of how matrix degradation is regulated and how myofibroblast cell fates can be manipulated, areas that may identify potential therapeutic approaches for fibrosis.
- Published
- 2018
8. Cellular senescence controls fibrosis in wound healing
- Author
-
Lester F. Lau and Joon Il Jun
- Subjects
Senescence ,Aging ,Pathology ,medicine.medical_specialty ,senescence ,integrin ,Integrin ,Biology ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Humans ,Myofibroblasts ,Cellular Senescence ,Cell Proliferation ,Skin ,030304 developmental biology ,Wound Healing ,0303 health sciences ,integumentary system ,Matricellular protein ,Cell Biology ,medicine.disease ,Extracellular Matrix ,Cell biology ,Oxidative Stress ,inflammation ,030220 oncology & carcinogenesis ,CYR61 ,Research Perspective ,biology.protein ,Wound healing ,CCN1 ,Myofibroblast ,Cysteine-Rich Protein 61 - Abstract
Mammalian wound healing involves the rapid synthesis and deposition of extracellular matrix (ECM) to maintain tissue integrity during repair. This process must be tightly controlled, as its deregulation may result in fibrosis, scarring, and loss of tissue function. Recent studies have uncovered an efficient and parsimonious mechanism for rendering fibrogenesis self-limiting in wound healing: in such diverse organs as the liver and skin, the myofibroblasts that initially proliferate and produce ECM are themselves eventually driven into senescence, blocking their further proliferation and converting them into matrix-degrading cells. Myofibroblast senescence in skin wounds is triggered by a dynamically expressed matricellular protein, CCN1/CYR61, which acts through integrin-mediated induction of oxidative stress. We propose that the onset of myofibroblast senescence is a programmed wound healing response that functions as a self-limiting mechanism for fibrogenesis, and this process may be regulated by the ECM microenvironment through the expression of CCN1/CYR61.
- Published
- 2010
- Full Text
- View/download PDF
9. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing
- Author
-
Lester F. Lau and Joon Il Jun
- Subjects
Senescence ,DNA damage ,Blotting, Western ,Biology ,Article ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,Humans ,Fibroblast ,Senolytic ,Cellular Senescence ,Cell Proliferation ,030304 developmental biology ,Wound Healing ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Matricellular protein ,Cell Biology ,Fibroblasts ,medicine.disease ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Microscopy, Fluorescence ,030220 oncology & carcinogenesis ,CYR61 ,RNA Interference ,Collagen ,Reactive Oxygen Species ,Wound healing ,Cysteine-Rich Protein 61 - Abstract
Cellular senescence is a recognized mechanism of tumour suppression; however, its contribution to other pathologies is not well understood. We show that the matricellular protein CCN1 (also known as CYR61; cysteine-rich protein 61), which is dynamically expressed at sites of wound repair, can induce fibroblast senescence by binding to integrin alpha(6)beta(1) and the heparan sulphate proteoglycans (receptors involved in cell adhesion). CCN1 induces DNA damage response pathways and activates p53 and the RAC1-NOX1 complex, which generates reactive oxygen species (ROS). This results in the ROS-dependent activation of the p16(INK4a)/pRb pathway, leading to senescence and concomitant expression of antifibrotic genes. Senescent fibroblasts accumulate in granulation tissues of healing cutaneous wounds and express antifibrotic genes in wild-type mice. These processes are lost in knockin mice that express a senescence-defective Ccn1 mutant, resulting in exacerbated fibrosis. Topical application of CCN1 protein to wounds reverses these defects. Thus, fibroblast senescence is a CCN1-dependent wound healing response in cutaneous injury that functions to curb fibrosis during tissue repair.
- Published
- 2010
- Full Text
- View/download PDF
10. Intracellular cleavage of osteopontin by caspase-8 modulates hypoxia/reoxygenation cell death through p53
- Author
-
Seon-Guk Choi, Chul Woong Chung, Hyung Ryong Kim, Joon Il Jun, In-Ki Kim, Hyun-Joo Kim, Han-Jung Chae, Yong-Keun Jung, Hyojin Kim, Yumin Oh, Il Sun Park, and Ho-June Lee
- Subjects
Caspase 8 ,Programmed cell death ,Multidisciplinary ,biology ,Blotting, Western ,Apoptosis ,Biological Sciences ,Molecular biology ,Cell Hypoxia ,stomatognathic system ,biology.protein ,Humans ,Osteopontin ,Viability assay ,Tumor Suppressor Protein p53 ,Protein kinase B ,Intracellular ,Caspase ,Densitometry ,HeLa Cells - Abstract
Osteopontin (OPN) is highly expressed in cancer patients and plays important roles in many stages of tumor progression, such as anti-apoptosis, proliferation, and metastasis. From functional screening of human cDNA library, we isolated OPN as a caspase-8 substrate that regulates cell death during hypoxia/reoxygenation (Hyp/RO). In vitro cleavage assays demonstrate that OPN is cleaved at Asp-135 and Asp-157 by caspase-8. Cellular cleavage of OPN is observed in apoptotic cells exposed to Hyp/RO among various apoptotic stimuli and its cleavage is blocked by zVAD or IETD caspase inhibitor. Further, over-expression of OPN, the form with secretion signal, inhibits Hyp/RO-induced cell death. Caspase cleavage-defective OPN mutant (OPN D135A/D157A) is more efficient to suppress Hyp/RO-induced cell death than wild-type OPN. OPN D135A/D157A sustains AKT activity to increase cell viability through inhibition of caspase-9 during Hyp/RO. In addition, OPN is highly induced in some tumor cells during Hyp/RO, such as HeLa and Huh-7 cells, which is associated with their resistance to Hyp/RO by sustaining AKT activity. Notably, OPN C-terminal cleavage fragment produced by caspase-8 is detected in the nucleus. Plasmid-encoded expression of OPN C-terminal cleavage fragment increases p53 protein level and induces apoptosis of wild-type mouse embryonic fibroblast cells, but not p53 −/− mouse embryonic fibroblast cells. These observations suggest that the protective function of OPN during Hyp/RO is inactivated via the proteolytic cleavage by caspase-8 and its cleavage product subsequently induces cell death via p53, postulating caspase-8 as a negative regulator of tumorigenic activity of OPN.
- Published
- 2009
- Full Text
- View/download PDF
11. The matricellular protein CCN1 mediates neutrophil efferocytosis in cutaneous wound healing
- Author
-
Lester F. Lau, Joon Il Jun, and Ki-Hyun Kim
- Subjects
Keratinocytes ,Integrins ,CCN1/CYR61 ,Neutrophils ,Integrin ,Fluorescent Antibody Technique ,General Physics and Astronomy ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Diabetes mellitus genetics ,Diabetes Mellitus ,Animals ,Humans ,Immunoprecipitation ,Receptors, Vitronectin ,Efferocytosis ,Opsonin ,Skin ,Wound Healing ,Multidisciplinary ,Neutrophil clearance ,Epidermal Growth Factor ,Reverse Transcriptase Polymerase Chain Reaction ,Macrophages ,Matricellular protein ,General Chemistry ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Cytophagocytosis ,Apoptosis ,Immunology ,biology.protein ,Receptors, Leptin ,Cell Migration Assays ,Wound healing ,Cysteine-Rich Protein 61 - Abstract
Neutrophil infiltration constitutes the first step in wound healing, although their timely clearance by macrophage engulfment, or efferocytosis, is critical for efficient tissue repair. However, the specific mechanism for neutrophil clearance in wound healing remains undefined. Here we uncover a key role for CCN1 in neutrophil efferocytosis by acting as a bridging molecule that binds phosphatidylserine, the 'eat-me' signal on apoptotic cells and integrins αvβ3/αvβ5 in macrophages to trigger efferocytosis. Both knockin mice expressing a mutant CCN1 that is unable to bind αvβ3/αvβ5 and mice with Ccn1 knockdown are defective in neutrophil efferocytosis, resulting in exuberant neutrophil accumulation and delayed healing. Treatment of wounds with CCN1 accelerates neutrophil clearance in both Ccn1 knockin mice and diabetic Lepr(db/db) mice, which suffer from neutrophil persistence and impaired healing. These findings establish CCN1 as a critical opsonin in skin injury and suggest a therapeutic potential for CCN1 in certain types of non-healing wounds.
- Published
- 2015
- Full Text
- View/download PDF
12. Essential Roles of Atg5 and FADD in Autophagic Cell Death
- Author
-
Dong-Hyung Cho, Yun Kyung Kwon, Ho-June Lee, Yong-Keun Jung, Joo Hang Kim, Joon Il Jun, Bo Youn Choi, Heuiran Lee, Yoshinori Oshumi, Noboru Mizushima, Jong Ok Pyo, Ha Na Woo, and Mi Hee Jang
- Subjects
Programmed cell death ,biology ,Autophagy ,ATG5 ,Cell Biology ,Vacuole ,Biochemistry ,Cell biology ,Cell culture ,biology.protein ,Ectopic expression ,FADD ,biological phenomena, cell phenomena, and immunity ,Molecular Biology ,Death domain - Abstract
Autophagic cell death is characterized by the accumulation of vacuoles in physiological and pathological conditions. However, its molecular event is unknown. Here, we show that Atg5, which is known to function in autophagy, contributes to autophagic cell death by interacting with Fas-associated protein with death domain (FADD). Down-regulation of Atg5 expression in HeLa cells suppresses cell death and vacuole formation induced by IFN-γ. Inversely, ectopic expression of Atg5 using adenoviral delivery induces autophagic cell death. Deletion mapping analysis indicates that procell death activity resides in the middle and C-terminal region of Atg5. Cells harboring the accumulated vacuoles triggered by IFN-γ or Atg5 expression become dead, and vacuole formation precedes cell death. 3-Methyladenine or expression of Atg5K130R mutant blocks both cell death and vacuole formation triggered by IFN-γ, whereas benzyloxycarbonyl-VAD-fluoromethyl ketone (Z-VAD-fmk) inhibits only cell death but not vacuole formation. Atg5 interacts with FADD via death domain in vitro and in vivo, and the Atg5-mediated cell death, but not vacuole formation, is blocked in FADD-deficient cells. These results suggest that Atg5 plays a crucial role in IFN-γ-induced autophagic cell death by interacting with FADD.
- Published
- 2005
- Full Text
- View/download PDF
13. Role of FLASH in caspase-8-mediated activation of NF-κB: dominant-negative function of FLASH mutant in NF-κB signaling pathway
- Author
-
Hyang-Sook Yoo, Jong Ok Pyo, Tae-Ho Lee, Ho-June Lee, Yong Sung Kim, Chul Woong Chung, Yong-Keun Jung, Nam-Soon Kim, Eunhee Kim, Joon Il Jun, and Kee Nyung Lee
- Subjects
Cancer Research ,TRAF2 ,Mutant ,Caspase 8 ,Cell Line ,Mice ,Genetics ,Animals ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Caspase ,Aspartic Acid ,biology ,Tumor Necrosis Factor-alpha ,Calcium-Binding Proteins ,HEK 293 cells ,NF-kappa B ,Transfection ,TNF Receptor-Associated Factor 2 ,NFKB1 ,Caspase Inhibitors ,Molecular biology ,Caspases ,Mutation ,biology.protein ,Death effector domain ,Apoptosis Regulatory Proteins ,Protein Binding ,Signal Transduction - Abstract
Caspase-8 is the most receptor-proximal, upstream caspase in the caspase cascade and plays a key role in cell death triggered by various death receptors. Here, we addressed the role of endogenous caspase-8 in tumor necrosis factor (TNF)-alpha-induced activation of NF-kappaB. Direct targeting of caspase-8 with siRNA and antisense (AS) approaches abolished TNF-alpha-induced activation of NF-kappaB in NIH3T3, HeLa, and HEK293 cells as determined with luciferase reporter gene and cell fractionation assays. Reconstitution of caspase-8-deficient C33A cells with processing-defective (P/D) mutant of caspase-8 sensitized the cells to TNF-alpha for NF-kappaB activation. In contrast to wild-type caspase-8, death effector domain mutant replacing Asp73 with Ala (caspase-8 (D73A)) failed to activate NF-kappaB and to bind FLICE-associated huge protein (FLASH) in vitro and in vivo. Instead, caspase-8 (D73A) mutant bound to caspase-8 and blocked NF-kappaB activation triggered by TNF-alpha and caspase-8. In addition, expression of an NF-kappaB-activating domain-deletion mutant of FLASH or transfection of FLASH AS oligonucleotides abolished TNF-alpha and caspase-8, but not phorbol 12-myristate 13-acetate, -induced activation of NF-kappaB. Further, immunoprecipitation assays showed that caspase-8 formed triple complex with TRAF2 and FLASH. Taken together, these results suggest that endogenous caspase-8 mediates TNF-alpha-induced activation of NF-kappaB via FLASH.
- Published
- 2004
- Full Text
- View/download PDF
14. Inhibition of Bcl10-mediated activation of NF-κB by BinCARD, a Bcl10-interacting CARD protein
- Author
-
Dong-Hyung Cho, Gil Sun Hong, Yong-Keun Jung, [No Value] Hyun-Woochoi, Jong Ok Pyo, Dong-Gyu Jo, Joon Il Jun, John Bertin, Ho-June Lee, In-Ki Kim, Chul Woong Chung, and Ha Na Woo
- Subjects
Transcription, Genetic ,T-Lymphocytes ,Amino Acid Motifs ,DNA Mutational Analysis ,HA ,Plasma protein binding ,Lymphocyte Activation ,Biochemistry ,Jurkat cells ,NF-κB ,Protein Structure, Secondary ,Conserved sequence ,Jurkat Cells ,Structural Biology ,Phosphorylation ,Luciferases ,Conserved Sequence ,Glutathione Transferase ,NF-kappa B ,Signal transducing adaptor protein ,Immunohistochemistry ,medicine.anatomical_structure ,Signal transduction ,Protein Binding ,Signal Transduction ,BinCARD ,Recombinant Fusion Proteins ,T cell ,Blotting, Western ,Molecular Sequence Data ,Biophysics ,Down-Regulation ,Biology ,Cell Line ,Leucine ,Two-Hybrid System Techniques ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,RNA, Messenger ,GST ,Molecular Biology ,Bcl10 ,Adaptor Proteins, Signal Transducing ,Sequence Homology, Amino Acid ,CARD ,Cell Biology ,B-Cell CLL-Lymphoma 10 Protein ,Precipitin Tests ,Molecular biology ,CARD Signaling Adaptor Proteins ,NF-κB activation ,Mutagenesis, Site-Directed ,Carrier Proteins ,HeLa Cells - Abstract
We have identified a novel CARD-containing protein from EST database. BinCARD (Bcl10-interacting protein with CARD). BinCARD was ubiquitously expressed. Co-immunoprecipitation, In vitro binding, mammalian two-hybrid, and immunostaining assays revealed that BinCARD interacted with Bcl10 through CARD. BinCARD potently suppressed NF-kappa B activation induced by Bcl10 and decreased the amounts of phosphorylated Bcl10. Mutations at the residue Leu17 or Leu65, which is highly conserved in CARD, abolished the inhibitory effects of BinCARD on both Bcl10-induced activation of NF-kappa B and phosphorylation of Bcl10. Further, expression of BinCARD inhibited Bcl10 phosphorylation induced by T cell activation signal. These results suggest that BinCARD interacts with Bcl10 to inhibit Bcl10-mediated activation of NF-kappa B and to suppress Bcl10 phosphorylation.
- Published
- 2004
- Full Text
- View/download PDF
15. Resolution of organ fibrosis.
- Author
-
Joon-Il Jun, Lau, Lester F., and Jun, Joon-Il
- Subjects
- *
FIBROSIS , *EXTRACELLULAR matrix , *TISSUE wounds , *WOUND healing , *GRANULATION tissue , *CYTOKINES - Abstract
Fibrosis is the excessive accumulation of extracellular matrix that often occurs as a wound healing response to repeated or chronic tissue injury, and may lead to the disruption of organ architecture and loss of function. Although fibrosis was previously thought to be irreversible, recent evidence indicates that certain circumstances permit the resolution of fibrosis when the underlying causes of injury are eradicated. The mechanism of fibrosis resolution encompasses degradation of the fibrotic extracellular matrix as well as elimination of fibrogenic myofibroblasts through their adaptation of various cell fates, including apoptosis, senescence, dedifferentiation, and reprogramming. In this Review, we discuss the present knowledge and gaps in our understanding of how matrix degradation is regulated and how myofibroblast cell fates can be manipulated, areas that may identify potential therapeutic approaches for fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
16. Essential roles of Atg5 and FADD in autophagic cell death: dissection of autophagic cell death into vacuole formation and cell death
- Author
-
Jong-Ok, Pyo, Mi-Hee, Jang, Yun-Kyung, Kwon, Ho-June, Lee, Joon-Il, Jun, Ha-Na, Woo, Dong-Hyung, Cho, Boyoun, Choi, Heuiran, Lee, Joo-Hang, Kim, Noboru, Mizushima, Yoshinori, Oshumi, and Yong-Keun, Jung
- Subjects
Binding Sites ,Cell Death ,Adenine ,Fas-Associated Death Domain Protein ,Gene Expression ,Saccharomyces cerevisiae ,Oligonucleotides, Antisense ,Transfection ,Amino Acid Chloromethyl Ketones ,Autophagy-Related Protein 5 ,Cell Line ,Interferon-gamma ,Mutagenesis ,Two-Hybrid System Techniques ,Vacuoles ,Autophagy ,Humans ,Microtubule-Associated Proteins ,Gene Deletion ,Adaptor Proteins, Signal Transducing ,HeLa Cells - Abstract
Autophagic cell death is characterized by the accumulation of vacuoles in physiological and pathological conditions. However, its molecular event is unknown. Here, we show that Atg5, which is known to function in autophagy, contributes to autophagic cell death by interacting with Fas-associated protein with death domain (FADD). Down-regulation of Atg5 expression in HeLa cells suppresses cell death and vacuole formation induced by IFN-gamma. Inversely, ectopic expression of Atg5 using adenoviral delivery induces autophagic cell death. Deletion mapping analysis indicates that procell death activity resides in the middle and C-terminal region of Atg5. Cells harboring the accumulated vacuoles triggered by IFN-gamma or Atg5 expression become dead, and vacuole formation precedes cell death. 3-Methyladenine or expression of Atg5(K130R) mutant blocks both cell death and vacuole formation triggered by IFN-gamma, whereas benzyloxycarbonyl-VAD-fluoromethyl ketone (Z-VAD-fmk) inhibits only cell death but not vacuole formation. Atg5 interacts with FADD via death domain in vitro and in vivo, and the Atg5-mediated cell death, but not vacuole formation, is blocked in FADD-deficient cells. These results suggest that Atg5 plays a crucial role in IFN-gamma-induced autophagic cell death by interacting with FADD.
- Published
- 2005
17. Calcium binding of ARC mediates regulation of caspase 8 and cell death
- Author
-
Dong-Hyung Cho, Joon Il Jun, Yong-Keun Jung, Yeon-Mi Hong, Do Han Kim, Dong-Gyu Jo, Chunghee Cho, Sang Mi Shim, Jae Woong Chang, Ho-June Lee, and Sungmin Song
- Subjects
Programmed cell death ,Caspase 2 ,Muscle Proteins ,Apoptosis ,In Vitro Techniques ,Caspase 8 ,Cell Line ,Jurkat Cells ,Animals ,Humans ,Molecular Biology ,Cell Growth and Development ,Caspase ,Death domain ,biology ,NLRP1 ,Cell Biology ,Recombinant Proteins ,Cell biology ,Enzyme Activation ,Caspases ,COS Cells ,biology.protein ,Thapsigargin ,Death effector domain ,Calcium ,Apoptosis Regulatory Proteins ,HeLa Cells - Abstract
Apoptosis or programmed cell death is genetically controlled and plays a central role in normal development and tissue homeostasis, including development of the nervous system and regulation of the immune system (8, 25, 35). Dysregulated apoptosis has been implicated in the pathogenesis of cancer and autoimmune, neurodegenerative, and cardiovascular diseases (28). The cell death machinery that is conserved throughout evolution is composed of activators, inhibitors, and effectors (14). The effector arm of the cell death pathway consists of a family of cysteinyl aspartate-specific proteases called caspases (2). Data suggest that apoptotic cell death can be brought about by the loss of Ca2+ homeostatic control, but it can also be finely tuned positively or negatively by more subtle changes in Ca2+ distribution within intracellular compartments (29). While protein kinases such as AKT and ERK have been reported to modulate caspase activity through phosphorylation (1, 7), there have been few regulatory molecules directly linking cytotoxic Ca2+ signaling to caspase activity. Based on sequence similarities, three prominent interaction motifs involved in apoptosis are recognized. The death domain superfamily consists of death domain (DD), death effector domain (DED), and caspase recruitment domain (CARD) families (16, 26). In recent years, a number of CARD-containing proteins have been identified and participate in various signaling pathways during apoptosis and NF-κB activation. ARC is a CARD protein that selectively interacts with the initiator caspase 8 and significantly attenuates death receptor-induced apoptosis (22). Recently, ARC was also found capable of blocking caspase-independent events such as hypoxia-induced cytochrome c release and hydrogen peroxide (H2O2)-induced necrotic cell death (10, 27). We also described the protective role of ARC during hypoxia of hippocampal neurons (17). In addition, ARC is known to be phosphorylated by protein kinase CK2, modulating the subcellular localization of ARC (23). While increasing evidence suggests an inhibitory role for ARC in the diverse cell death processes, the precise mechanism by which ARC interferes with caspase-dependent and caspase-independent cell death has not been defined yet. In the present study, we postulate that ARC is a Ca2+-binding CARD protein that modulates activation of caspase 8.
- Published
- 2004
18. Fas-associated factor 1, FAF1, is a member of Fas death-inducing signaling complex
- Author
-
Seung-Wook Ryu, Soojin Lee, Min Young Park, Joon Il Jun, Eunhee Kim, and Yong-Keun Jung
- Subjects
Programmed cell death ,Death Domain Receptor Signaling Adaptor Proteins ,Fas-Associated Death Domain Protein ,Molecular Sequence Data ,Apoptosis ,Biochemistry ,Jurkat cells ,Fas ligand ,Receptors, Tumor Necrosis Factor ,Jurkat Cells ,Humans ,FADD ,Amino Acid Sequence ,Molecular Biology ,Death domain ,Adaptor Proteins, Signal Transducing ,Caspase 8 ,biology ,Cell Biology ,Fas receptor ,Caspase 9 ,Cell biology ,Protein Structure, Tertiary ,Caspases ,Death-inducing signaling complex ,biology.protein ,Apoptosis Regulatory Proteins ,Carrier Proteins ,Sequence Alignment ,Signal Transduction - Abstract
FAF1 has been introduced as a Fas-binding protein. However, the function of FAF1 in apoptotic execution is not established. Based on the fact that FAF1 is a Fas-binding protein, we asked if FAF1 interacted with other members of the Fas-death-inducing signaling complex (Fas-DISC) such as Fas-associated death domain protein (FADD) and caspase-8. FAF1 could interact with caspase-8 and FADD in vivo as well as in vitro. The death effector domains (DEDs) of caspase-8 and FADD interacted with the amino acid 181–381 region of FAF1, previously known to have apoptotic potential. Considering that FAF1 directly binds to Fas and caspase-8, FAF1 shows similar protein-interacting characteristics to that of FADD. In the coimmunoprecipitation with an anti-Fas antibody (APO-1) in Jurkat cells, endogenous FAF1 was associated with the precipitates in which caspase-8 was present. By confocal microscopic analysis, both Fas and FAF1 were detected in the cytoplasmic membrane before Fas activation, and in the cytoplasm after Fas activation. FADD and caspase-8 colocalized with Fas in Jurkat cells validating the presence of FAF1 in the authentic Fas-DISC. Overexpression of FAF1 in Jurkat cells caused significant apoptotic death. In addition, the FAF1 deletion mutant lacking the N terminus where Fas, FADD, and caspase-8 interact protected Jurkat cells from Fas-induced apoptosis demonstrating dominant-negative phenotype. Cell death by overexpression of FAF1 was suppressed significantly in both FADD- and caspase-8-deficient Jurkat cells when compared with that in their parental Jurkat cells. Collectively, our data show that FAF1 is a member of Fas-DISC acting upstream of caspase-8.
- Published
- 2003
19. Erratum: The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing
- Author
-
Joon Il Jun and Lester F. Lau
- Subjects
Senescence ,medicine.anatomical_structure ,Fibrosis ,Matricellular protein ,medicine ,Cell Biology ,Cutaneous wound ,Biology ,Fibroblast ,medicine.disease ,Cell biology - Published
- 2010
- Full Text
- View/download PDF
20. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing.
- Author
-
Joon-Il Jun and Lau, Lester F.
- Subjects
- *
CARRIER proteins , *CELLULAR aging , *TUMOR growth , *FIBROSIS , *PROTEOGLYCANS , *CELL receptors , *CELL adhesion - Abstract
Cellular senescence is a recognized mechanism of tumour suppression; however, its contribution to other pathologies is not well understood. We show that the matricellular protein CCN1 (also known as CYR61; cysteine-rich protein 61), which is dynamically expressed at sites of wound repair, can induce fibroblast senescence by binding to integrin α6β1 and the heparan sulphate proteoglycans (receptors involved in cell adhesion). CCN1 induces DNA damage response pathways and activates p53 and the RAC1–NOX1 complex, which generates reactive oxygen species (ROS). This results in the ROS-dependent activation of the p16INK4a/pRb pathway, leading to senescence and concomitant expression of antifibrotic genes. Senescent fibroblasts accumulate in granulation tissues of healing cutaneous wounds and express antifibrotic genes in wild-type mice. These processes are lost in knockin mice that express a senescence-defective Ccn1 mutant, resulting in exacerbated fibrosis. Topical application of CCN1 protein to wounds reverses these defects. Thus, fibroblast senescence is a CCN1-dependent wound healing response in cutaneous injury that functions to curb fibrosis during tissue repair. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
21. Intracellular cleavage of osteopontin by caspase-8 modulates hypoxia/reoxygenation cell death through p53.
- Author
-
Hyo-Jin Kim, Ho-June Lee, Joon-Il Jun, Yumin Oh, Seon-Guk Choi, Hyunjoo Kim, Chul-Woong Chung, In-Ki Kim, Il-Sun Park, Han-Jung Chae, Hyung-Ryong Kim, and Yong-Keun Jung
- Subjects
OSTEOPONTIN ,CANCER invasiveness ,CELL death ,P53 antioncogene ,BIOLOGICAL assay ,APOPTOSIS ,FIBROBLASTS ,THERAPEUTICS - Abstract
Osteopontin (OPN) is highty expressed in cancer patients and plays important roles in many stages of tumor progression, such as antiapoptosis. proliferation, and metastasis. From functional screening of human cDNA library, we isolated OPN as a caspase-8 substrate that regulates cell death during hypoxia/reoxygenation (Hyp/RO). In vitro cleavage assays demonstrate that OPN is cleaved at Asp-135 and Asp-157 by caspase-8. Cellular cleavage of OPN is observed in apoptotic cells exposed to Hyp/RO among various apoptotic stimuli and its cleavage is blocked by zVAD or IETD caspase inhibitor. Further, over-expression of OPN, the form with secretion signal, inhibits Hyp/RO-induced cell death. Caspase cleavage-defective OPN mutant (OPN D135A/D157A) is more efficient to suppress Hyp/RO-induced cell death than wild-type OPN. OPN D135A/D1S7A sustains AKT activity to increase cell viability through inhibition of caspase-9 during Hyp/RO. In addition, OPN is highly induced in some tumor cells during Hyp/RO, such as HeLa and Huh-7 cells, which is associated with their resistance to Hyp/RO by sustaining AKT activity. Notably, OPN Cterminal cleavage fragment produced by caspase-8 is detected in the nucleus. Plasmid-encoded expression of OPN C-terminal cleavage fragment increases p53 protein level and induces apoptosis of wildtype mouse embryonic fibroblast cells, but not p53
-/- mouse embryonic fibroblast cells. These observations suggest that the protective function of OPN during Hyp/RO is inactivated via the proteolytic cleavage by caspase-8 and its cleavage product subsequently induces cell death via p53, postulating caspase-8 as a negative regulator of tumorigenic activity of OPN. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
22. Fas-associated 1, FAF1, Is a Member of Fas Death-inducing Signaling Complex.
- Author
-
Seung-Wook Ryu, Soo-Jin Lee, Min-Young Park, Joon-il Jun, Yong-Keun Jung, and Eunhee Kim
- Subjects
- *
PROTEINS , *APOPTOSIS - Abstract
Studies the function of Fas-associated factor 1 (FAF1) protein in apoptotic execution. Interaction of FAF1 with fas-death inducing signaling complex members; Interaction between FAF1 and caspase-8; Death effector domains of FAF1.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.