• Title/Summary/Keyword: Caspase-3 protease

Search Result 77, Processing Time 0.033 seconds

A Caspase Inducing Inhibitor Isolated from Caesalpinia sappan (소목(Caesalpinia sappan)으로부터 분리한 caspase 유도 저해 물질)

  • Son, Eun-Jung;Kim, Jin-Hee;Kim, Hyun-A;Baek, Seung-Hwa;Kho, Yung-Hee;Kim, Mee-Ree;Lee, Choong-Hwan
    • Korean Journal of Food Science and Technology
    • /
    • v.35 no.4
    • /
    • pp.680-683
    • /
    • 2003
  • Through the screening of caspase-3 inducing inhibitors in U937 human monocytic leukemia cell from natural sources, Caesalpiniae sappan, which showed a high level of inhibition, was selected. The inhibition compound was purified from methanol extract by silica gel column chromatography and HPLC. The inhibitor was identified as brazilin by spectroscopic methods of ESI-MS, $^1H-NMR$, and $^{13}C-NMR$. Brazilin showed inhibitory activity of caspase-3 induction, a major protease of apoptosis cascade, with $IC_{50}$ value of $4.5\;{\mu}g/mL$ after 7 hr of treatment in U937 cells.

Protective Effects of Hwansodan(Huanshao-dan) Water Extract in Serum Deprivation-induced Apoptosis of PC12 Cells (환소단이 영양혈청 결핍성 PC12 신경세포의 apoptosis에 미치는 영향)

  • 임준식;김명선;소홍섭;이지현;한상혁;허윤;박래길;문병순
    • The Journal of Korean Medicine
    • /
    • v.21 no.4
    • /
    • pp.64-72
    • /
    • 2000
  • Objectives : This study was designed to investigate the neuroprotective effect of Hwansodan(Huanshao-dan) on the apoptosis induced by withdrawal of neurotrophic support. Methods : PCl2 pheochromocytoma cells have been used extensively as a model for studying the cellular and molecular effects of neuronal cells. The viability of cells was measured by MTT assay. We used DNA fragmentation and caspase 3-like protease activation assay. Results : The water extract of Hwansodan(Huanshao-dan) significantly showed protective effects on serum and glucose deprivation-induced apoptotic death. Hwansodan(Huanshao-dan) also prevents DNA fragmentation and caspase 3-like protease activation, representing typical neuronal apoptotic phenomena in PCl2 pheochromocytoma cells and induces tyrosine phosphorylation of proteins around 44 kDa, which was identified as ERK1 with electrophoretic gel mobility shift by Western blot. In addition, MAPK kinase(MEK) inhibitor PD98059 and Ras inactivator, ${\alpha}-hydroxyfarnesylphosphonic$ acid attenuated the neuroprotective effects of Hwansodan(Huanshao-dan) in serum-deprived PCl2 cells. Conclusions : These results indicate that Ras/MEK/ERK signaling pathway plays a key role in neuroprotective effects of Hwansodan(Huanshao-dan) in serum-deprived PCl2 cells. Taken together, we suggest the possibility that Hwansodan(Huanshao-dan) might provide a neurotrophic-like activity in PCl2 cells.

  • PDF

Inhibitory Effect of Apoptosis of Human Astrocytes by Juniper Oil (신경교(神經膠) 성상세포(星狀細胞)에서 쥬니퍼오일에 의한 세포자멸사(細胞自滅死)의 억제(抑制) 효과(效果))

  • Kim Tae-Heong;Kim Tae-Heon;Lee Sung-Ryull;Lyu Yeoung-Su
    • Journal of Oriental Neuropsychiatry
    • /
    • v.11 no.2
    • /
    • pp.1-9
    • /
    • 2000
  • In previous studies, heat shock has been reported to induce the apoptosis or programmed cell death through the activation of caspase-3. 1 investigated the effect of juniper pure essential oil on the heat shock-induced apoptosis in human astrocyte cell line CCF-STTGI. Treatment of the astrocytes with heat shock markedly induced apoptotic cell death. However, pretreatment of the astrocytes with juniper oil ingibited the heat shock-induced apoptosis. To determine whether juniper inhibits the heat shock-induced activation of these apoptotic proteases, activation of CPP32 was assessed by Western blotting. Consistent with flow cytometry. DNA fragmentation and giemsa staining, heat shock-induced activation of CPP32 was blocked by juniper oil. Poly(ADP-ribose) polymerase (PARP), cysteine protease substrates were fragmented as a consequence of apoptosis by heat shock. Juniper oil inhibited the PARP fragmentation. This juniper oil also inhibited the heat shock-induced activation of caspase-3. These results suggest that juniper oil may modulate the apoptosis through the activation of the interleukin-1-converting enzyme-like protease.

  • PDF

Role of p-38 MAP Kinase in apoptosis of hypoxia-induced osteoblasts (저산소 상태로 인한 조골세포 고사사기전에서 p-38 MAP kinase의 역할에 관한 연구)

  • Yoon, Jeong-Hyeon;Jeong, Ae-Jin;Kang, Kyung-Hwa;Kim, Sang-Cheol
    • The korean journal of orthodontics
    • /
    • v.33 no.3 s.98
    • /
    • pp.169-183
    • /
    • 2003
  • Tooth movement by orthodontic force effects great tissue changes within the periodontium, especially by shifting the blood flow in the pressure side and resulting in a hypoxic state of low oxygen tension. The aim of this study is to elucidate the possible mechanism of apoptosis in response to hypoxia in MC3T3El osteoblasts, the main cells in bone remodeling during orthodontic tooth movement. MC3T3El osteoblasts under hypoxic conditions ($2\%$ orygen) resulted in apoptosis in a time-dependent manner as estimated by DNA fragmentation assay and nuclear morphology stained with fluorescent dye, Hoechst 33258. Pretreatment with Z-VAD-FMK, a pancaspase inhibitor, or Z-DEVD-CHO, a specific caspase-3 inhibitor, completely suppressed the DNA ladder in response to hypoxia. An increase in caspase-3-like protease (DEVDase) activity was observed during apoptosis, but no caspase-1 activity (YVADase) was detected. To confirm what caspases are involved in apoptosis, Western blot analysis was performed using anti-caspase-3 or -6 antibodies. The 10-kDa protein, corresponding to the active products of caspase-3, and the 10-kDa protein of the active protein of caspase-6 were generated in hypoxia-challenged cells in which the processing of the full length form of caspase-3 and -6 was evident. While a time course similar to this caspase-3 and -6 activation was evident, hypoxic stress caused the cleavage of lamin A, which was typical of caspase-6 activity. In addition, the stress elicited the release of cytochrome c into the cytosol during apoptosis. Furthermore, we observed that pre-treatment with SB203580, a selective p38 mitogen activated protein kinase inhibitor, attenuated the hypoxia-induced apoptosis. The addition of SB203S80 suppressed caspase-3 and -6-like protease activity by hypoxia up to $50\%$. In contrast, PD98059 had no effect on the hypoxia-induced apoptosis. To confirm the involvement of MAP kinase, JNK/SAPK, ERK, or p38 kinase assay was performed. Although p38 MAPK was activated in response to hypoxic treatment, the other MAPK -JNK/SAPK or ERK- was either only modestly activated or not at all. These results suggest that p38 MAPK is involved in hypoxia-induced apoptosis in MC3T3El osteoblasts.

Apoptosis Induction by Menadione in Human Promyelocytic Leukemia HL-60 Cells

  • Sa, Duck-Jin;Lee, Eun-Jee;Yoo, Byung-Sun
    • Toxicological Research
    • /
    • v.25 no.3
    • /
    • pp.113-118
    • /
    • 2009
  • Cell death induced by menadione (vitamin K-3,2-methyl-1,4-naphthoquinone) has been investigated in human promyelocytic leukemia HL-60 cells. Menadione was found to induce both apoptosis and necrosis in HL-60 cells. Low concentration ($1{\sim}$50 ${\mu}$M) of menadione induced apoptotic cell death, which was demonstrated by typical DNA ladder patterns on agarose gel electrophoresis and flow cytometry analysis. In contrast, a high concentration of menadione (100 ${\mu}$M) induced necrotic cell death, which was demonstrated by DNA smear pattern in agarose gel electrophoresis. Necrotic cell death was accompanied with a great reduction of cell viability. Menadione activated caspase-3, as evidenced by both increased protease activity and proteolytic cleavage of 116 kDa poly(ADP-ribose) polymerase (PARP) into 85 kDa cleavage product. Caspase-3 activity was maximum at 50 ${\mu}$M of menadione, and very low at 100 ${\mu}$M of menadione. Taken together, our results showed that menadione induced mixed types of cell death, apoptosis at low concentrations and necrosis at high concentrations in HL-60 cells.

Study on Synergistic Anti-tumor Effect of Combination with Adriamycin and Palginhonhapwhajucwhan (팔진탕합화적환(八珍湯合化積丸)과 Adriamycin의 병용처리시 나타나는 synergistic 항종양(抗腫瘍) 효과(效果)에 관(關)한 작용기전 연구(硏究))

  • Moon, Gu;Moon, Seok-Jae;Won, Jin-Hee;Cho, Jung-Yun;Park, Sang-Gu;Song, Bong-Gil;Park, Rae-Gil;Lee, Byung-Gu
    • The Journal of Internal Korean Medicine
    • /
    • v.21 no.3
    • /
    • pp.443-452
    • /
    • 2000
  • Objective : This study was designed to evaluate the synergistic effect on cytotoxicity of combination with adriamycin and Palginhonhapwhajucwhan, a traditional prescription for cancer treatment in oriental medicine, in Chang, HL-60, Hep-3B and Alexander cells. Methods : We observed cell viability in Chang, HL-60, Hep-3B, and Alexander cells by crystal violet staining. Those cells were treated with various concentrations of adriamycin alone, Palginhonhapwhajucwhan alone and combination of two medications for 10 hr. On condition of $0.5{\mu}l/ml$ adriamycin alone, $15.6{\mu}l/ml$ Paljintanghapwhajucwhan alone and combination of two medications, at first, we observed colony forming of Chang and HL-60 cells. Second, we observed DNA fragmentation by agarose electrophoresis in Chang, HL-60, Hep-38 and Alexander cells. Third, we measured the catalytic activation of caspase-1, 2, 3, 6, 8, and 9 protease in Chang cells and caspase-3 protease in Chang, HL-60, Hep-3B and Alexander cells by using fluorogenic substrate. Finally, we isolated mRNA of Fas in Chang, HL-60, Hep-38 and Alexander cells and observed that Fas gene was amplified by RT-PCR Results : 1. The combination of adriamycin and Palginhonhapwhajucwhan synergistically augmented the cytotoxicity of Chang and HL-60 cells whereas did not in Hep-38 and Alexander cells. 2. Cotreatment of two drugs also markedly inhibited the colony forming ability both in Chang and HL-60 cells. 3. The cytotoxicity of these medicatons was revealed as apoptosis characterized by high molecular wight DNA fragmentaton. 4. The apoptotic cytotoxicity was mediated by activation of caspase-3 protease in Chang cells. 5. Synergistic increase in apoptotic cytotoxicity by combination of two medications was dependent on the expression of Fas in cancer cells. Conclusions : Combination of adriamycin and Palginhonhapwhajucwhan significantly augmented apoptotic cytotoxicity of Fas-positive cells such as Chang and HL-60 cells via acticaton of apoptosis signaling pathway.

  • PDF

Study on Anti-Cancer Effects of Backhapgogumtanggami-bang (백합고금탕가미방의 항종양 효과에 관한 연구)

  • 김병주;문구;문석재;원진희;김태균;배남규
    • The Journal of Korean Medicine
    • /
    • v.22 no.2
    • /
    • pp.64-74
    • /
    • 2001
  • Objectives : The effects of aqueous extract of Backhapgogumtanggami-bang (BGTG, a newly devised herb medicine) on the induction of apoptotic cell death were investigated in human lymphoid origin leukemia cell lines, HL-60. Methods : Cells were treated with various concentrations and $400{\;}\mu\textrm{g}/ml$ BGTG for 12 hr. Genomic DNA was isolated and separated on 1.8% agarose gels. Lysates from the cells were used to measure the activity of caspase-2, -3, -8, and -9 protease by using fluorogenic peptide. Cells were preincubated with SB-203580 for 30 min. Nuclear protein from the cells was incubated with oliginucleotide probe of AP-l and NF-kB. Nuclear extracts from the cells were isolated and reacted with antibodies. Results : The viability of HL-60 cells were markedly decreased by BGTG extract in a dose- and time-dependent manner. BGTG extract induced the apoptotic death of HL-60 cells which was characterized by the DNA fragmentation. The activations of Caspase-2, 3, and 9 were induced by BGTG. However, selective inhibition of the p38 mitogen-activated protein kinase pathways by SB-203580 did not affect the extent of BGTG extract-induced cell death. Furthermore, we observed the transient activations of transcriptional factors such as AP-l and NF-kB. Conclusions : These results suggest that BGTG extract induced apoptotic death of HL-60 cells and caspase activations as well as the modulation of transcriptional factors such as AP-1 and NF-kB.

  • PDF

Roles of ginsenosides in inflammasome activation

  • Yi, Young-Su
    • Journal of Ginseng Research
    • /
    • v.43 no.2
    • /
    • pp.172-178
    • /
    • 2019
  • Inflammation is an innate immune response that protects the body from pathogens, toxins, and other dangers and is initiated by recognizing pathogen-associated molecular patterns or danger-associated molecular patterns by pattern-recognition receptors expressing on or in immune cells. Intracellular pattern-recognition receptors, including nucleotide-binding oligomerization domain-like receptors (NLRs), absent in melanoma 2, and cysteine aspartate-specific protease (caspase)-4/5/11 recognize various pathogen-associated molecular patterns and danger-associated molecular patterns and assemble protein complexes called "inflammasomes." These complexes induce inflammatory responses by activating a downstream effector, caspase-1, leading to gasdermin D-mediated pyroptosis and the secretion of proinflammatory cytokines, such as interleukin $(IL)-1{\beta}$ and IL-18. Ginsenosides are natural steroid glycosides and triterpene saponins found exclusively in the plant genus Panax. Various ginsenosides have been identified, and their abilities to regulate inflammatory responses have been evaluated. These studies have suggested a link between ginsenosides and inflammasome activation in inflammatory responses. Some types of ginsenosides, including Rh1, Rg3, Rb1, compound K, chikusetsu saponin IVa, Rg5, and Rg1, have been clearly demonstrated to inhibit inflammatory responses by suppressing the activation of various inflammasomes, including the NLRP3, NLRP1, and absent in melanoma 2 inflammasomes. Ginsenosides have also been shown to inhibit caspase-1 and to decrease the expression of $IL-1{\beta}$ and IL-18. Given this body of evidence, the functional relationship between ginsenosides and inflammasome activation provides new insight into the understanding of the molecular mechanisms of ginsenoside-mediated antiinflammatory actions. This relationship also has applications regarding the development of antiinflammatory remedies by ginsenoside-mediated targeting of inflammasomes, which could be used to prevent and treat inflammatory diseases.

Inducing Apoptosis of NCI-H157 Human Lung Carcinoma Cells via Activation of Caspase Cascade by Combination Treatment with Arsenic Trioxide and Sulindac (NCI-H157 폐암 세포주에서 Caspase Cascade 활성을 통한 Arsenic Trioxide와 Sulindac 병합요법의 세포고사효과)

  • Kim, Hak Ryul;Yang, Sei Hoon;Jeong, Eun Taik
    • Tuberculosis and Respiratory Diseases
    • /
    • v.56 no.4
    • /
    • pp.381-392
    • /
    • 2004
  • Arsenic trioxide($As_2O_3$) was introduced into the treatment of refractory or relapsed acute promyelocytic Ieukemia. Some investigators have reported that arsenic trioxide had induced apoptosis in a variety of solid human tumor cell lines, including non-small cell lung cancer. Non-steroidal anti-inflammatory drugs(NSAIDs) are powerful chemopreventive agents for gastrointestinal cancers and the growth of established tumors are reduced by inducing apoptosis. It's also reported that NSAIDs enhanced tumor response to chemotherapeutic drugs or radiation. In this study, we aimed to determine whether combination of arsenic trioxide with sulindac augmented its apoptotic potential in NCI-H157 human lung cancer cells. The human lung cancer cell line NCI-H157 was treated with arsenic trioxide and sulindac. Cell viability was measured by the MTT assay. Apoptosis was measured by nuclear staining and flow cytometric analysis. The catalytic activity of the caspase families were measured by the fluorogenic cleavage of biosubstrates. The western blotting were also performed to define the mechanical basis of apoptosis. Combination treatment of arsenic trioxide and sulindac decreased the viability of NCI-H157 human lung cancer cells in a dose-dependent manner. The catalytic activity of caspase-3, 8 and 9 proteases were increased after combination treatment. Consistently PARP was cleaved from 116kDa to 85kDa fragments, and the expression of ICAD was decreased by time-dependent manner. Also combination treatment increased the expression of Fas and Fas/L. Combination therapy of arsenic trioxide with sulindac augments cell death and induces apoptosis via the activation of caspase cascade in NCI-H157 human lung carcinoma cells.