• Title/Summary/Keyword: Heme-oxygenase 1

Search Result 354, Processing Time 0.032 seconds

Neuroprotective Effect of Ethyl Acetate Fraction of Portulaca oleracea L. (마치현 에틸아세테이트 분획물의 뇌세포 보호효과)

  • Im, Nam Kyung;Jeong, Gil Saeng
    • Korean Journal of Pharmacognosy
    • /
    • v.44 no.4
    • /
    • pp.379-383
    • /
    • 2013
  • Portulaca oleracea L. is known to have many biological benefits such as anti-oxidant, anti-inflammatory, anti-allergic and anti-tumor. The objective of this study is to explore the neuroprotective effect of P. oleracea L. against glutamate-induced oxidative stress in mouse hippocampal HT22 cells. P. oleracea L. 70% ethanol extract and solvent fractions have the potent neroprotective effects on glutamate-induced nerotoxicity by induced the expression of heme oxygenase (HO)-1 in HT22 cells. Especially, ethyl acetate fraction showed higher protective effect. In HT22 cell, P. oleracea L. treatment with ERK inhibitor (PD98059) and c-JUN N-terminal kinase (JNK) inhibitor (SP600125) reduced P. oleracea L. ethyl acetate fraction induced HO-1 expression and P. oleracea L. ethyl acetate fraction also increased ERK and JNK phosphorylation. Furthermore, we found that treatment of P. oleracea L. caused the nuclear accumulation of Nrf2. In conclusion, the ethyl acetate fraction of 70% ethanol extract of P. oleracea L. significantly protect glutamate-induced oxidative damage by induction of HO-1 via Nrf2, ERK and JNK pathway in mouse hippocampal HT22. Taken together these finding suggest that P. oleracea L. ethyl acetate fraction is good source for taking active compounds and may be a potential therapeutic agent for brain disorder that induced by oxidative stress and neuronal damage.

Effects of Allium hookeri Extracts on Glutamate-induced Neurotoxicity in HT22 Cells (글루타메이트로 유발한 세포독성에 대한 삼채추출물의 뇌세포 보호 효과)

  • Kim, Ji-Yun;Ko, Wonmin;Kim, Ae-Jung
    • Korean Journal of Pharmacognosy
    • /
    • v.48 no.1
    • /
    • pp.31-37
    • /
    • 2017
  • Glutamate-induced oxidative stress results in neuro-degenerative disorders in many central nervous system (CNS) such as Alzheimer's disease, ischemia, Huntington's disease, and Parkinson's disease. Our study was performed to investigate neuroprotective effects of Allium hookeri extracts (leaf, root, and whole) on glutamate-induced HT22 cells. In this study, ethanol extract of A. hookeri showed the outstanding neuroprotective effect in HT22 cells. In addition, we found that ethanol extract of A. hookeri root increased heme oxygenase (HO)-1 in HT22 cells. Moreover, ethanol extract of A. hookeri root also upregulated nuclear accumulation of nuclear factor E2-related factor 2 (Nrf2) in HT22 cells. These results demonstrate that ethanol extract of A. hookeri root contributes neuroprotective effects against glutamate-induced oxidative stress in HT22 cells, via Nrf2-mediated HO-1 expression. Our study suggests that ethanol extract of A. hookeri root could be the potential agent for the treatment of many neuro-degenerative diseases.

Neuroprotective Effects of Ethanol Extract of Ganoderma lucidum L. on murine hippocampal cells (영지 에탄올 추출물의 마우스 유래 뇌 해마세포 보호효과)

  • Lee, Seung Cheol;Im, Nam-Kyung;Jeong, Hye Young;Choi, Eun Hwa;Jeon, Soo Myeong;Jeong, Gil-Saeng
    • Korean Journal of Pharmacognosy
    • /
    • v.45 no.2
    • /
    • pp.161-167
    • /
    • 2014
  • Ganoderma lucidum L. (GL) is a traditional oriental medicine that has been widely used as anti-inflammatory, antitumor and anti-oxidant in Korea and other Asian countries. In this study, we investigated the ethanol extract of GL has neuroprotective effects in murine hippocampal HT22 cells. GL ethanol extract has the potent neuroprotective effects on glutamate-intoxicated cells by inducing the expression of heme oxygenase (HO)-1 in HT22 cells. GL ethanol extract increased JNK phosphorylation. Obviously, When we treated the GL extract with c-Jun N-terminal kinase (JNK) inhibitor (SP600125), HO-1 expression was reduced. Moreover, we found that GL treatment caused the nuclear accumulation of Nrf2. In conclusion, the ethanol extract of GL significantly protects glutamate-induced oxidative damage by induction of HO-1 via Nrf2, JNK pathway in mouse hippocampal HT22. These results suggest that GL ethanol extract would be a good source for taking active compounds and may be a potential pharmaceutical products for brain disorder induced by neuronal damage and oxidative stress.

A Formulated Korean Red Ginseng Extract Inhibited Nitric Oxide Production through Akt- and Mitogen Activated Protein Kinase-dependent Heme Oxygenase-1 Upregulation in Lipoteichoic Acid-stimulated Microglial Cells (홍삼추출액은 lipoteichoic acid로 자극된 소교세포에서 Akt 및 MAPK 의존적으로 heme oxygenase-1 발현을 유도함으로써 NO 생성을 억제함)

  • Shin, Ji Eun;Lee, Kyungmin;Kim, Ji-Hee;Madhi, Iskander;Kim, YoungHee
    • Journal of Life Science
    • /
    • v.29 no.4
    • /
    • pp.402-409
    • /
    • 2019
  • Korean red ginseng made from steaming and drying fresh ginseng has long been used as a traditional herbal medicine due to its effects on the immune, endocrine, and central nerve systems and its anti-inflammatory activity. In this study, we investigated the molecular mechanism responsible for the anti-inflammatory effects of a formulated Korean red ginseng extract (RGE) in response to lipoteichoic acid (LTA), a cell wall component of gram-positive bacteria. RGE inhibited LTA-induced nitric oxide (NO) secretion and inducible nitric oxide synthase (iNOS) expression in BV-2 microglial cells, without affecting cell viability. RGE also inhibited nuclear translocation of nuclear factor kappa B ($NF-{\kappa}B$) p65 and degradation of $I{\kappa}B-{\alpha}$. In addition, RGE increased the expression of heme oxygenase-1 (HO-1) in a dose-dependent manner, and the inhibitory effect of RGE on iNOS expression was abrogated by small interfering RNA-mediated knockdown of HO-1. Moreover, RGE induced nuclear translocation of nuclear factor E2-related factor 2 (Nrf2), a transcription factor that regulates HO-1 expression. Furthermore, the phosphoinositide-3-kinase (PI-3K) inhibitor and mitogen-activated protein kinase (MAPK) inhibitors suppressed RGE-mediated expression of HO-1, and RGE enhanced the phosphorylation of Akt, extracellular signal-regulated kinases (ERKs), p38, and c-JUN N-terminal kinases (JNKs). These results suggested that RGE suppressed the production of NO, a proinflammatory mediator, by inducing HO-1 expression via PI-3K/Akt- and MAPK-dependent signaling in LTA-stimulated microglia. The findings indicate that RGE could be used for the treatment of neuroinflammation induced by grampositive bacteria and that it may have therapeutic potential for various neuroinflammation-associated disorders.

Resveratrol Ameliorates NMDA-induced Mitochondrial Injury by Enhanced Expression of Heme Oxygenase-1 in HT-22 Neuronal Cells (NMDA를 처리한 HT-22 신경세포에서 미토콘드리아 손상을 완화하는 레스베라트롤의 보호 효과와 헴 산화효소-1의 역할)

  • Kang, Jae Hoon;Woo, Jae Suk
    • Journal of Life Science
    • /
    • v.32 no.1
    • /
    • pp.11-22
    • /
    • 2022
  • N-methyl-D-aspartate (NMDA) receptors have received considerable attention regarding their involvement in glutamate-induced neuronal excitotoxicity. Resveratrol has been shown to exhibit neuroprotective effects against this kind of overactivation, but the underlying cellular mechanisms are not yet clearly understood. In this study, HT-22 neuronal cells were treated with NMDA in Mg2+-free buffer and subsequently used as an experimental model of glutamate excitotoxicity to elucidate the mechanisms of resveratrol-induced neuroprotection. We found that NMDA treatment causes a drop in MTT reduction ability, disrupts inside-negative transmembrane potential of mitochondria, depletes cellular ATP levels, and stimulates intracellular ROS production. Double fluorescence imaging studies demonstrated an increased formation of mitochondrial permeability transition (MPT) pores accompanied by apoptotic cell death, while cobalt protoporphyrin and bilirubin showed protective effects against NMDA-induced mitochondrial injury. On the other hand, zinc protoporphyrin IX significantly attenuated the protective effects of resveratrol which was itself shown to enhance heme oxygenase-1 (HO-1) mRNA and protein expression levels. In cells transfected with HO-1 small interfering RNA, resveratrol failed to suppress the NMDA-induced effects on MTT reduction ability and MPT pore formation. The present study suggests that resveratrol may prevent mitochondrial injury in NMDA- treated HT-22 cells and that enhanced expression of HO-1 is involved in the underlying cellular mechanism.

Protective effect of p53 in vascular smooth muscle cells against nitric oxide-induced apoptosis is mediated by up-regulation of heme oxygenase-2

  • Kim, Young-Myeong;Choi, Byung-Min;Kim, Yong-Seok;Kwon, Young-Guen;Kibbe, Melina R.;Billiar, Timothy R.;Tzeng, Edith
    • BMB Reports
    • /
    • v.41 no.2
    • /
    • pp.164-169
    • /
    • 2008
  • The tumor suppressor gene p53 regulates apoptotic cell death and the cell cycle. In this study, we investigated the role of p53 in nitric oxide (NO)-induced apoptosis in vascular smooth muscle cells (VSMCs). We found that the NO donor S-nitroso-N-acetyl-penicillamine (SNAP) increased apoptotic cell death in p53-deficient VSMCs compared with wild-type cells. The heme oxygen-ase (HO) inhibitor tin protoporphyrin IX reduced the resistance of wild-type VSMCs to SNAP-induced cell death. SNAP promoted HO-1 expression in both cell types. HO-2 protein was increased only in wild-type VSMCs following SNAP treatment; however, similar levels of HO-2 mRNA were detected in both cell types. SNAP significantly increased the levels of non-heme-iron and dinitrosyl iron-sulfur clusters in wild-type VSMCs compared with p53-deficient VSMCs. Moreover, pretreatment with FeSO4 and the carbon monoxide donor CORM-2, but not biliverdin, significantly protected p53-deficient cells from SNAP-induced cell death compared with normal cells. These results suggest that wild-type VSMCs are more resistant to NO-mediated apoptosis than p53-deficient VSMCs through p53-dependent up-regulation of HO-2.

Capsaicin Ameliorates Cisplatin-Induced Renal Injury through Induction of Heme Oxygenase-1

  • Jung, Sung-Hyun;Kim, Hyung-Jin;Oh, Gi-Su;Shen, AiHua;Lee, Subin;Choe, Seong-Kyu;Park, Raekil;So, Hong-Seob
    • Molecules and Cells
    • /
    • v.37 no.3
    • /
    • pp.234-240
    • /
    • 2014
  • Cisplatin is one of the most potent chemotherapy agents. However, its use is limited due to its toxicity in normal tissues, including the kidney and ear. In particular, nephrotoxicity induced by cisplatin is closely associated with oxidative stress and inflammation. Heme oxygenase-1(HO-1), the rate-limiting enzyme in the heme metabolism, has been implicated in a various cellular processes, such as inflammatory injury and anti-oxidant/oxidant homeostasis. Capsaicin is reported to have therapeutic potential in cisplatin-induced renal failures. However, the mechanisms underlying its protective effects on cisplatin-induced nephrotoxicity remain largely unknown. Herein, we demonstrated that administration of capsaicin ameliorates cisplatin-induced renal dysfunction by assessing the levels of serum creatinine and blood urea nitrogen (BUN) as well as tissue histology. In addition, capsaicin treatment attenuates the expression of inflammatory mediators and oxidative stress markers for renal damage. We also found that capsaicin induces HO-1 expression in kidney tissues and HK-2 cells. Notably, the protective effects of capsaicin were completely abrogated by treatment with either the HO inhibitor ZnPP IX or HO-1 knockdown in HK-2 cells. These results suggest that capsaicin has protective effects against cisplatin-induced renal dysfunction through induction of HO-1 as well as inhibition oxidative stress and inflammation.

Effect of Chlorella vulgaris Intake on Antioxidative Capacity in Rats Oxidatively Stressed with Dietary Cadmium

  • Kim, You-Jin;Jeong, Se-Won;Kwon, Sang-Hee;Kim, Mi-Kyung
    • Food Science and Biotechnology
    • /
    • v.18 no.5
    • /
    • pp.1055-1062
    • /
    • 2009
  • This study was conducted to investigate whether dietary chlorella intake could have an effect on antioxidative capacity in rats oxidatively stressed with cadmium (Cd). Sprague-Dawley rats fed dietary chlorella (0, 5, and 10%) for 4 weeks after induction of oxidative stress by exposing to Cd (200 ppm) for 8 weeks. After the oxidative stress applied, plasma and liver malondialdehyde concentrations and xanthine oxidase activities were decreased in 5% chlorella fed group compared to chlorella free group. Although liver heme oxygenase-1 protein expression was not affected by chlorella, the enzyme activity was improved in 5% chlorella fed group. Erythrocyte superoxide dismutase activity and hepatic metallothionein concentration were increased in 5% chlorella fed group. However, 10% chlorella intake had no effect on the improvement of oxidative stress-related enzymes and proteins. These findings suggest that, after induction of oxidative stress with Cd, 5% chlorella intake might improve antioxidative capacity against oxidative stress.

Involvement of Peroxynitrite in NO Donor-Induced HO-1 Expression in Rat Articular Chondrocytes (흰쥐 관절연골세포에서 NO donor에 의해 유도된 HO-1 발현에서 peroxynitrite의 관련성 연구)

  • Song, Ju-Dong;Kim, Kang-Mi;Kim, Jong-Min;Yoo, Young-Hyun;Park, Young-Chul
    • Journal of Life Science
    • /
    • v.21 no.4
    • /
    • pp.486-493
    • /
    • 2011
  • Nitric oxide (NO) donors are a potent inducer of heme oxygenase-1 (HO-1). However, it is unclear whether or not HO-1 expression induced by NO donors is a direct consequence of NO released by NO donors. Here, we investigated the effects of NO donors on the expression of HO-1 in primary rat articular chondrocytes. NO donors (SIN-1, SNAP, and SNP) significantly induced the accumulation of HO-1 protein accompanied by an increase in HO-1 mRNA. NO donor-induced HO-1 expression exerted cytoprotection against NO and/or superoxide-induced cell death. Guanylate cyclase signaling was not associated with Nrf2 and HO-1 expression in NO donor-treated chondrocytes. Interestingly, NO scavenger carboxy-PTIO and SOD mimetic TEMPOL markedly inhibited NO donor-induced HO-1 expression in chondrocytes. In addition, NO donor-induced HO-1 expression was completely abrogated by the peroxynitrite scavenger MnTBAP. Since peroxynitrite can be physiologcally formed in the cell through reaction of NO with superoxide, we analyzed whether or not peroxynitrite could directly induce HO-1 expression in chondrocytes. Peroxynitrite treatment in chondrocytes evoked doseand time-dependent Nrf2 and HO-1 expression. These results indicate that HO-1 expression induced by NO donors in rat articular chondrocytes is due to NO-mediated peroxynitrite rather than NO.

A Novel Pyrazolo[3,4-d]pyrimidine Induces Heme Oxygenase-1 and Exerts Anti-Inflammatory and Neuroprotective Effects

  • Lee, Ji Ae;Kwon, Young-Won;Kim, Hye Ri;Shin, Nari;Son, Hyo Jin;Cheong, Chan Seong;Kim, Dong Jin;Hwang, Onyou
    • Molecules and Cells
    • /
    • v.45 no.3
    • /
    • pp.134-147
    • /
    • 2022
  • The anti-oxidant enzyme heme oxygenase-1 (HO-1) is known to exert anti-inflammatory effects. From a library of pyrazolo[3,4-d]pyrimidines, we identified a novel compound KKC080096 that upregulated HO-1 at the mRNA and protein levels in microglial BV-2 cells. KKC080096 exhibited anti-inflammatory effects via suppressing nitric oxide, interleukin1β (IL-1β), and iNOS production in lipopolysaccharide (LPS)-challenged cells. It inhibited the phosphorylation of IKK and MAP kinases (p38, JNK, ERK), which trigger inflammatory signaling, and whose activities are inhibited by HO-1. Further, KKC080096 upregulated anti-inflammatory marker (Arg1, YM1, CD206, IL-10, transforming growth factor-β [TGF-β]) expression. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridinetreated mice, KKC080096 lowered microglial activation, protected the nigral dopaminergic neurons, and nigral damage-associated motor deficits. Next, we elucidated the mechanisms by which KKC080096 upregulated HO-1. KKC080096 induced the phosphorylation of AMPK and its known upstream kinases LKB1 and CaMKKbeta, and pharmacological inhibition of AMPK activity reduced the effects of KKC080096 on HO-1 expression and LPS-induced NO generation, suggesting that KKC080096-induced HO-1 upregulation involves LKB1/AMPK and CaMKKbeta/AMPK pathway activation. Further, KKC080096 caused an increase in cellular Nrf2 level, bound to Keap1 (Nrf2 inhibitor protein) with high affinity, and blocked Keap1-Nrf2 interaction. This Nrf2 activation resulted in concurrent induction of HO-1 and other Nrf2-targeted antioxidant enzymes in BV-2 and in dopaminergic CATH.a cells. These results indicate that KKC080096 is a potential therapeutic for oxidative stress-and inflammation-related neurodegenerative disorders such as Parkinson's disease.