• 제목/요약/키워드: Cilostazol

검색결과 29건 처리시간 0.032초

렛트를 이용한 심적환과 cilostazol에 관한 상호작용 연구 (Pharmacokinetics Interaction between Cardiotonic Pills and Cilostazol in Rats)

  • 김익균
    • 생명과학회지
    • /
    • 제26권1호
    • /
    • pp.123-128
    • /
    • 2016
  • 본 연구의 최종 목표는 한방제제인 심적환과 심혈관계 양방 치료제인 cilostazol과 병용 투여 효과에 대한 포괄적이고 통합의학 측면에서 정확한 정보를 얻는 것이다. Cilostazol은 말초 동맥질환 치료제로 개발 된 항 혈소판 및 혈관확장제이다. 사이클릭 AMP protein kinase A를 활성 시켜 세포내 사이클릭 AMP (cAMP) 증가를 통하여 내피세포의 NO생산을 활성화 시킨다. 심적환을 단 회 또는 반복 투여 후 cilostazol의 약물 동태학적 효과를 평가하기 위하여 순수한 증류수 단회 용량과 증류수에 심적환 콜로이드 현탁액을 각각 대조군과 시험군에 투여 하여 30분 후, 두 그룹에 cilostazol를 투여하였다. 혈청은 cilostazol 약물 투여 30분 전에 수집 하였으며, cilostazol 약물 처리 후 0.25, 0.5, 0.45 및 1, 2, 4, 6, 8, 24시간 후에 각각 수집 하였다. 그 다음 실험군과 및 테스트 그룹 사이에 실로 스타 졸에서 관찰 된 약동학 적 변화를 평가 하였다. 통계적으로 유의 한 차이는 심적관 단독 투여와 반복투여군 그룹의 약물 동태 학에서 관찰되지 않았다. 이러한 연구 결과는 만성 질환 환자에서 한약제인 심적환의 투여는 cilostazol의 약동학에 영향을 미치지 않았음을 보여 주었다. 본 연구에서 얻어진 결과는 만성 혈관질환 환자에서 심적환과 cilostazol의 병용 투여를 제안하며 두 약물간의 잠재적 인 약물 상호 작용에 대한 cilostazol의 생체 이용률에 영향을 미치지 않을 것이라 판단된다.

Cilostazol에 의한 뇌혈관내피세포의 세포이동 증진 효과연구 (Cilostazol Promotes the Migration of Brain Microvascular Endothelial Cells)

  • 이세원;박정화;신화경
    • 생명과학회지
    • /
    • 제26권12호
    • /
    • pp.1367-1375
    • /
    • 2016
  • Cilostazol은 phosphodiesterase III의 선택적 저해제로 알려져 있으며, 뇌졸중 치료에 일반적으로 사용되고 있다. Cilostazol을 처리한 경우, 국소 뇌허혈이 발생한 후에 혈관신생을 통해서 혈관형성이 향상된다는 것을 본 연구자들이 발표하였다. 혈관신생은 조직의 허혈상태를 극복하기 위해서 혈관재생을 촉진하는 중요한 과정으로써, 혈관내피세포의 증식, 이동, 모세관구조 형성의 다단계 과정으로 구성되어 있다. 이에 본 연구에서는 인간 뇌혈관내피세포를 이용하여 cilostazol이 혈관신생의 각 단계들에 어떤 영향을 미치는지 조사하였다. Cilostazol은 농도의존적으로 뇌혈관내피세포의 이동성을 촉진하였으나, 뇌혈관내피세포의 증식과 모세관구조 형성에는 영향을 미치지 않았다. Cilostazol이 세포이동을 조절하는 기전을 분석하기 위해서 cDNA microarray를 수행하였고, 세포이동에 관련성이 있는 5종의 후보 유전자들을 선택하여 real-time PCR을 통해 해당 유전자의 발현을 검증하였다. Cilostazol에 의해서 발현양이 조절되는 유전자들로써, phosphoserine aminotransferase 1 (PSAT1)와 CCAAT/enhancer binding protein ${\beta}$ ($C/EBP{\beta}$)은 발현이 증가하였고, tissue factor pathway inhibitor 2 (TFPI2), retinoic acid receptor responder 1 (RARRES1), RARRES3는 발현이 감소하였다. 이상의 결과를 통해서 cilostazol이 혈관내피세포의 이동을 촉진하여 혈관신생을 향상시킬 수 있음을 제안할 수 있으며, 뇌혈관내피세포에 대한 cilostazol의 조절기전에 대해서 더욱 상세히 규명을 한다면 혈관형성을 통하여 허혈성 질환을 치료할 수 있는 유용한 정보가 될 것으로 기대한다.

Cilostazol Decreases Ethanol-Mediated TNFalpha Expression in RAW264.7 Murine Macrophage and in Liver from Binge Drinking Mice

  • Lee, Youn-Ju;Eun, Jong-Ryeol
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제16권2호
    • /
    • pp.131-138
    • /
    • 2012
  • Alcoholic hepatitis is a leading cause of liver failure in which the increased production of tumor necrosis factor ${\alpha}$ (TNF${\alpha}$) plays a critical role in progression of alcoholic liver disease. In the present study, we investigated the effects of cilostazol, a selective inhibitor of type III phosphodiesterase on ethanol-mediated TNF${\alpha}$ production in vitro and $in$ $vivo$, and the effect of cilostazol was compared with that of pentoxifylline, which is currently used in clinical trial. RAW264.7 murine macrophages were pretreated with ethanol in the presence or absence of cilostazol then, stimulated with lipopolysacchride (LPS). Cilostazol significantly suppressed the level of LPS-stimulated TNF${\alpha}$ mRNA and protein with a similar degree to that by pentoxifylline. Cilostazol increased the basal AMP- activated protein kinase (AMPK) activity as well as normalized the decreased AMPK by LPS. AICAR, an AMPK activator and db-cAMP also significantly decreased TNF${\alpha}$ production in RAW264.7 cells, but cilostazol did not affect the levels of intracellular cAMP and reactive oxygen species (ROS) production. The $in$ $vivo$ effect of cilostazol was examined using ethanol binge drinking (6 g/kg) mice model. TNF${\alpha}$ mRNA and protein decreased in liver from ethanol gavaged mice compared to that from control mice. Pretreatment of mice with cilostazol or pentoxifylline further reduced the TNF${\alpha}$ production in liver. These results demonstrated that cilostazol effectively decrease the ethanol-mediated TNF${\alpha}$ production both in murine macrophage and in liver from binge drinking mice and AMPK may be responsible for the inhibition of TNF${\alpha}$ production by cilostazol.

Negligible Effect of Ginkgo Biloba Extract on the Pharmacokinetics of Cilostazol

  • Chung, Hye-Jin;Kim, Nam-Sun;Kim, Eun-Jeong;Kim, Tae-Kon;Ryu, Keun-Ho;Lee, Bong-Yong;Kim, Dong-Hyun;Jin, Chang-Bae;Yoo, Hye-Hyun
    • Biomolecules & Therapeutics
    • /
    • 제17권3호
    • /
    • pp.311-317
    • /
    • 2009
  • Ginkgo biloba (G. biloba) extract is a widely used phytomedicine for the oral treatment of peripheral vascular disease. Cilostazol is a synthetic antiplatelet and vasodilating agent for the treatment of intermittent claudication resulting from peripheral arterial disease. It is likely to use concomitantly G. biloba extract and cilostazol for the treatment of peripheral arterial disease, which raises a concern of increasing their adverse effects of herbal-drug interactions. To clarify any possible herbal-drug interaction between G. biloba extract and cilostazol, the effect of the G. biloba extract on the pharmacokinetics of cilostazol was investigated. As cilostazol is known to be eliminated mainly by cytochrome P450 (CYP)-mediated metabolism, we investigated the effects of G. biloba extract on the human CYP enzyme activities and the effect of G. biloba extract on the pharmacokinetics of cilostazol after co-administration of the two agents to male beagle dogs. The G. biloba extract inhibited more or less CYP2C8, CYP2C9, and CYP2C19 enzyme activities in the in vitro microsomal study with $IC_{50}$ values of 30.8, 60.5, and $25.2{\mu}g/ml$, respectively. In the pharmacokinetic study, co-administration with the G. biloba extract had no significant effect on the pharmacokinetics of cilostazol in dogs, although CYP2C has been reported to be responsible for the metabolism of cilostazol. In conclusion, these results suggest that there may not be a pharmacokinetic interaction between G. biloba extract and cilostazol.

프레탈 정(실로스타졸 100 mg)에 대한 엘지실로스타졸 정의 생물학적 동등성 (Bioequivalence of LG Cilostazol Tablet to Pletaal Tablet (Cilostazol 100 mg))

  • 조혜영;임동구;신상철;문재동;이용복
    • 한국임상약학회지
    • /
    • 제11권1호
    • /
    • pp.7-12
    • /
    • 2001
  • Cilostazol has both antithrombotic and cerebral vasodilating effects, and one of the mechanism is the selective inhibition of platalet cyclic AMP phosphodiesterase. Bioequivalence of two cilostazol tablets, the $Pletaal^{TM}$ (Korea Otsuka Pharmaceutical Co.) and the LG $Cilostazol^{TM}$ (LG Chemical Co.), was evaluated according to the guidelines of Korea Food and Drug Administration (KFDA). Sixteen normal male volunteers ($20\sim29$ years old) were randomly divided into two groups and a randomized $2\times2$ cross-over study was employed. After oral administration of $Pletaal^{TM}$ or LG $Cilostazol^{TM}$ tablet (100 mg cilostazol), blood samples were taken at predetermined time intervals and the serum cilostazol concentrations were determined using an HPLC method with UV/VIS detector. The pharmacokinetic parameters $(AUC_t,\;C_{max}\;and\;T_{max})$ were calculated and ANOVA was utilized for the statistical analysis. The results showed that the differences in AUCt, C_{max} and Tmax between two tablets based on the $Pletaal^{TM}$ tablet were $-5.39\%,\;2.32\%\;and\;4.26\%$, respectively. The powers (1-${\beta}$) for $AUC_t,\;C_{max}\;and\;T_{max}\;were\;83.81\%,\;96.02\%\;and\;91.04%$, respectively. Minimum detectable differences ($\Delta$) and $90\%$ confidence intervals were all less than $\pm20\%$. All these parameters met the criteria of KFDA for bioequivalence, indicating that LG $Cilostazol^{TM}$ tablet is bioequivalent to $Pletaal^{TM}$ tablet.

  • PDF

Cilostazol Inhibits Vascular Smooth Muscle Cell Proliferation and Reactive Oxygen Species Production through Activation of AMP-activated Protein Kinase Induced by Heme Oxygenase-1

  • Kim, Jung-Eun;Sung, Jin-Young;Woo, Chang-Hoon;Kang, Young-Jin;Lee, Kwang-Youn;Kim, Hee-Sun;Kwun, Woo-Hyung;Choi, Hyoung-Chul
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제15권4호
    • /
    • pp.203-210
    • /
    • 2011
  • Cilostazol is a selective inhibitor of phosphodiesterase 3 that increases intracellular cAMP levels and activates protein kinase A, thereby inhibiting vascular smooth muscle cell (VSMC) proliferation. We investigated whether AMP-activated protein kinase (AMPK) activation induced by heme oxygenase-1 (HO-1) is a mediator of the beneficial effects of cilostazol and whether cilostazol may prevent cell proliferation and reactive oxygen species (ROS) production by activating AMPK in VSMC. In the present study, we investigated VSMC with various concentrations of cilostazol. Treatment with cilostazol increased HO-1 expression and phosphorylation of AMPK in a dose- and time-dependent manner. Cilostazol also significantly decreased platelet-derived growth factor (PDGF)-induced VSMC proliferation and ROS production by activating AMPK induced by HO-1. Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production.

HO-1 Induced by Cilostazol Protects Against TNF-${\alpha}$-associated Cytotoxicity via a PPAR-${\gamma}$-dependent Pathway in Human Endothelial Cells

  • Park, So-Youn;Bae, Jin-Ung;Hong, Ki-Whan;Kim, Chi-Dae
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제15권2호
    • /
    • pp.83-88
    • /
    • 2011
  • A large body of evidence has indicated that induction of endogenous antioxidative proteins seems to be a reasonable strategy for delaying the progression of cell injury. In our previous study, cilostazol was found to increase the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in synovial cells. Thus, the present study was undertaken to examine whether cilostazol is able to counteract tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$)-induced cell death in endothelial cells via the induction of HO-1 expression. We exposed human umbilical vein endothelial cells (HUVECs) to TNF-${\alpha}$ (50 ng/ml), with or without cilostazol ($10{\mu}M$). Pretreatment with cilostazol markedly reduced TNF-${\alpha}$-induced viability loss in the HUVECs, which was reversed by zinc protoporphyrine IX (ZnPP), an inhibitor of HO-1. Moreover, cilostazol increased HO-1 protein and mRNA expression. Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). In an assay measuring peroxisome proliferator-activated receptor-${\gamma}$ (PPAR-${\gamma}$) transcription activity, cilostazol directly increased PPAR-${\gamma}$ transcriptional activity which was completely abolished by HO-1 inhibitor. Furthermore, increased PPAR-${\gamma}$ activity by cilostazol and rosiglitazone was completely abolished in cells transfected with HO-1 siRNA. Taken together, these results indicate that cilostazol up-regulates HO-1 and protects cells against TNF-${\alpha}$-induced endothelial cytotoxicity via a PPAR-${\gamma}$-dependent pathway.

Cilostazol attenuates kainic acid-induced hippocampal cell death

  • Park, Young-Seop;Jin, Zhen;Jeong, Eun Ae;Yi, Chin-ok;Lee, Jong Youl;Park, In Sung;Roh, Gu Seob
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제22권1호
    • /
    • pp.63-70
    • /
    • 2018
  • Cilostazol is a selective inhibitor of type 3 phosphodiesterase (PDE3) and has been widely used as an antiplatelet agent. Cilostazol mediates this activity through effects on the cyclic adenosine monophosphate (cAMP) signaling cascade. Recently, it has attracted attention as a neuroprotective agent. However, little is known about cilostazol's effect on excitotoxicity induced neuronal cell death. Therefore, this study evaluated the neuroprotective effect of cilostazol treatment against hippocampal neuronal damage in a mouse model of kainic acid (KA)-induced neuronal loss. Cilostazol pretreatment reduced KA-induced seizure scores and hippocampal neuron death. In addition, cilostazol pretreatment increased cAMP response element-binding protein (CREB) phosphorylation and decreased neuroinflammation. These observations suggest that cilostazol may have beneficial therapeutic effects on seizure activity and other neurological diseases associated with excitotoxicity.

Amyloid β에 의해 유도된 신경세포 손상에 대한 phosphodiesterase III inhibitor인 cilostazol의 신경보호 효과 (Phosphodiesterase III Inhibitor Cilostazol Protects Amyloid β-Induced Neuronal Cell Injury via Peroxisome Proliferator-Activated Receptor-γ Activation)

  • 박선행;김지현;배순식;홍기환;최병태;신화경
    • 생명과학회지
    • /
    • 제21권5호
    • /
    • pp.647-655
    • /
    • 2011
  • Amyloid ${\beta}$ ($A{\beta}$)의 신경독성은 알츠하이머병의 주된 원인이 되고 이러한 신경독성은 일련의 신경세포 사멸반응에 의해 일어난다고 알려져 있다. 본 연구에서는 알츠하이머병의 실험모델로 mouse primary neuronal cell에 $A{\beta}_{25-35}$를 처리하여 세포독성을 유도하는 세포실험모델과 C57BL/6J mouse 뇌실에 $A{\beta}_{25-35}$를 주입하여 인지장애를 일으키는 동물실험모델을 이용하여 phosphodiesterase III 억제제인 cilostazol의 신경보호 효과에 대해 조사하였다. $A{\beta}_{25-35}$를 신경세포에 처리하면 세포생존율이 감소되었고, 세포사멸이 일어난 세포의 수도 증가되었다. 이러한 $A{\beta}_{25-35}$에 의한 세포독성이 cilostazol처리에 의해 회복되었으며, peroxisome proliferator-activated receptor(PPAR)-${\gamma}$ 항진제인 rosiglitazone 또한 동일한 회복효과를 나타내었다. Cilostazol과 rosiglitazone에 의한 이러한 회복효과가 PPAR-${\gamma}$ 길항제인 GW9662에 의해 다시 억제되는 결과를 통해 cilostazol의 효과는 PPAR-${\gamma}$가 매개하는 신호전달이 관여함을 알 수 있었다. 직접 PPAR-${\gamma}$ 활성화 정도를 측정한 결과, $A{\beta}_{25-35}$ 처리에 의해 감소된 PPAR-${\gamma}$ 활성화 정도가 cilostazol과 rosiglitazone에 의해 증가함을 관찰할 수 있었고, 이는 GW9662에 의해 다시 억제됨을 확인하였다. 게다가, cilostazol은 세포사멸이 일어난 세포의 수와 세포사멸 조절단백질인 Bax/Bcl-2의 비율도 감소시켰다. Cilostazol (20 mg/kg, 구강투여)을 C57BL/6J mice 뇌실에 $A{\beta}_{25-35}$를 주입하기 2주 동안 전처리하고, $A{\beta}_{25-35}$ 주입 후 4주 동안 처리하면, 기억력과 학습능력을 증진시킨다는 결과를 water maze 실험을 통해 알 수 있었으며, rosiglitazone (10 mg/kg)을 먹인 동물에서도 동일한 결과를 얻을 수 있었다. 본 연구를 통해서 cilostazol이 PPAR-${\gamma}$ 활성화를 통해 $A{\beta}_{25-35}$로 인한 신경세포 손상과 세포사멸을 약화시켜, 신경세포의 생존을 증진시키고, 알츠하이머에서 인지장애를 개선할 것으로 생각된다. 따라서, phosphodiesterase III 억제제인 cilostazol은 알츠하이머 질병 치료에 새로운 전략이 될 수 있을 것이다.

Synergistic Efficacy of Concurrent Treatment with Cilostazol and Probucol on the Suppression of Reactive Oxygen Species and Inflammatory Markers in Cultured Human Coronary Artery Endothelial Cells

  • Park, So-Youn;Lee, Jeong-Hyun;Shin, Hwa-Kyoung;Kim, Chi-Dae;Lee, Won-Suk;Rhim, Byung-Yong;Shin, Yung-Woo;Hong, Ki-Whan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제12권4호
    • /
    • pp.165-170
    • /
    • 2008
  • In the present study, we aimed to identify the synergistic effects of concurrent treatment of low concentrations of cilostazol and probucol to inhibit the oxidative stress with suppression of inflammatory markers in the cultured human coronary artery endothelial cells (HCAECs). Combination of cilostazol (0.3${\sim}3{\mu}$M) with probucol (0.03${\sim}0.3{\mu}$M) significantly suppressed TNF-${\alpha}$-stimulated NAD(P)H-dependent superoxide, lipopolysaccharide (LPS)-induced intracellular reactive oxygen species (ROS) production and TNF-${\alpha}$ release in comparison with probucol or cilostazol alone. The combination of cilostazol (0.3${\sim}3{\mu}$M) with probucol (0.1${\sim}0.3{\mu}$M) inhibited the expression of vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) more significantly than did the monotherapy with either probucol or cilostazol. In line with these results, combination therapy significantly suppressed monocyte adhesion to endothelial cells. Taken together, it is suggested that the synergistic effectiveness of the combination therapy with cilostazol and probucol may provide a beneficial therapeutic window in preventing atherosclerosis and protecting from cerebral ischemic injury.