• Title/Summary/Keyword: Rapamycin

Search Result 179, Processing Time 0.029 seconds

Rapamycin Inhibits Rabbit Corneal Neovascularization Induced by Angiogenin (Rapamycin의 angiogenin 유도성 가토 각막의 혈관신생 억제)

  • 권영삼;김재찬;장광호
    • Journal of Veterinary Clinics
    • /
    • v.21 no.3
    • /
    • pp.309-313
    • /
    • 2004
  • The purpose of this study was to determine whether immunosuppresant, rapamycin could inhibit corneal angiogenesis induced by angiogenin and to evalutate the its role by micropocket assay. The rabbit's eye was implanted intrastromally into the superior cornea with pellet for the control group, pellet containing of angiogenin for the angiogenin group, and pellet containing of angiogenin and rapamycin for the rapamycin group. We could observed that the angiogen induced corneal angiogenesis was inhibited by rapamycin. The score of neovascularization was significantly decreased in the rapamycin group than in the angiogenin group at 7 and 10 days after pellet implantation (p < 0.05). Histologically, the cornea treated with rapamycin group also showed much less new vessels than the cornea treated with angiogenin. In conclusion, rapamycin appears to inhibit angiogenin induced angiogenesis in a rabbit corneal micropocket assay and may have therapeutic potential as an antiangiogenic agent.

Metabolomic Response of Chlamydomonas reinhardtii to the Inhibition of Target of Rapamycin (TOR) by Rapamycin

  • Lee, Do Yup;Fiehn, Oliver
    • Journal of Microbiology and Biotechnology
    • /
    • v.23 no.7
    • /
    • pp.923-931
    • /
    • 2013
  • Rapamycin, known as an inhibitor of Target of Rapamycin (TOR), is an immunosuppressant drug used to prevent rejection in organ transplantation. Despite the close association of the TOR signaling cascade with various scopes of metabolism, it has not yet been thoroughly investigated at the metabolome level. In our current study, we applied mass spectrometric analysis for profiling primary metabolism in order to capture the responsive dynamics of the Chlamydomonas metabolome to the inhibition of TOR by rapamycin. Accordingly, we identified the impact of the rapamycin treatment at the level of metabolomic phenotypes that were clearly distinguished by multivariate statistical analysis. Pathway analysis pinpointed that inactivation of the TCA cycle was accompanied by the inhibition of cellular growth. Relative to the constant suppression of the TCA cycle, most amino acids were significantly increased in a time-dependent manner by longer exposure to rapamycin treatment, after an initial down-regulation at the early stage of exposure. Finally, we explored the isolation of the responsive metabolic factors into the rapamycin treatment and the culture duration, respectively.

Induced Autophagy Regulates Salmonella enterica serovar Typhimurium Infection in Murine Macrophage (쥐의 큰포식세포주에서 자가포식현상에 의한 Salmonella enterica serovar Typhimurium의 감염 조절)

  • Lee, Sunhye;Kim, Ju-Young;Lee, Hyo-Ji;Jung, Yu-Jin
    • Korean Journal of Microbiology
    • /
    • v.50 no.1
    • /
    • pp.27-32
    • /
    • 2014
  • Autophagy is one of the lysosomal degradation pathways to maintain cellular homeostasis. The damaged proteins or organelles are uptaken through extra- and intra-cellular stress, starvation and infected pathogens, subsequently, autophagosomes are fused with lysosomes to break down the molecules. Salmonella enterica serovar Typhimurium (S. Typhimurium), intracellular bacteria, cause acute gastroenteritis and food poisoning. Given that autophagy induced by S. Typhimurium plays an important role in the cells to control the infection, we identify whether the induction of autophagy with rapamycin, chemical inducer of autophagy, before infection regulates S. Typhimurium infection. After treatment of rapamycin or 3-methyladenine (3-MA), autophagy inhibitor, RAW264.7 cells were infected with S. Typhimurium. Pretretment of rapamycin decreased the growth rate of S. Typhimurium in the cells; otherwise, pretreatment of 3-MA increased the growth rate of S. Typhimurium. The expression of autophagy-related genes was significantly increased in the S. Typhimurium-infected cells pretreated with rapamycin. To examine whether induced autophagy by rapamycin control the infection with increase the production of reactive oxygen species (ROS) and nitric oxide (NO), antibacterial radical substrates were measured in infected cells followed by the treatment with either rapamycin or 3-MA. NO production increased in RAW264.7 cells; otherwise, ROS production remained unchanged during the infection. These findings suggest that inducing autophagy with rapamycin reveals antimicrobial activity as producing NO against S. Typhimurium infection in mouse macrophages.

Genomewide Profiling of Rapamycin Sensitivity in Saccharomyces cerevisiae on Synthetic Medium

  • Chang, Yeon-Ji;Shin, Chun-Shik;Han, Dong-Hun;Kim, Ji-Yun;Kim, Kang-In;Kwon, Yong-Min;Huh, Won-Ki
    • Genomics & Informatics
    • /
    • v.8 no.4
    • /
    • pp.177-184
    • /
    • 2010
  • The target of rapamycin (TOR) signaling pathway is a conserved pathway that regulates eukaryotic cell growth in response to environmental cues. Chemical genomic approaches that profile rapamycin sensitivity of yeast deletion strains have given insights into the function of TOR signaling pathway. In the present study, we analyzed the rapamycin sensitivity of yeast deletion library strains on synthetic medium. As a result, we identified 130 strains that are hypersensitive or resistant to rapamycin compared with wild-type cells. Among them, 36 genes are newly identified to be related to rapamycin sensitivity. Moreover, we found 16 strains that show alteration in rapamycin sensitivity between complex and synthetic media. We suggest that these genes may be involved in part of TOR signaling activities that is differentially regulated by media composition.

Rapamycin Influences the Efficiency of In vitro Fertilization and Development in the Mouse: A Role for Autophagic Activation

  • Lee, Geun-Kyung;Shin, Hyejin;Lim, Hyunjung Jade
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.29 no.8
    • /
    • pp.1102-1110
    • /
    • 2016
  • The mammalian target of rapamycin (mTOR) regulates cellular processes such as cell growth, metabolism, transcription, translation, and autophagy. Rapamycin is a selective inhibitor of mTOR, and induces autophagy in various systems. Autophagy contributes to clearance and recycling of macromolecules and organelles in response to stress. We previously reported that vitrified-warmed mouse oocytes show acute increases in autophagy during warming, and suggested that it is a natural response to cold stress. In this follow-up study, we examined whether the modulation of autophagy influences survival, fertilization, and developmental rates of vitrified-warmed mouse oocytes. We used rapamycin to enhance autophagy in metaphase II (MII) oocytes before and after vitrification. The oocytes were then subjected to in vitro fertilization (IVF). The fertilization and developmental rates of vitrified-warmed oocytes after rapamycin treatment were significantly lower than those for control groups. Modulation of autophagy with rapamycin treatment shows that rapamycin-induced autophagy exerts a negative influence on fertilization and development of vitrified-warmed oocytes.

Effects of Glycerol and Shikimic Acid on Rapamycin Production in Streptomyces rapamycinicus

  • La, Huyen Thi Huong;Nguyen, Thao Kim Nu;Dinh, Hang Thuy;Nguyen, Quyen Minh Huynh;Nguyen, Minh Hong
    • Microbiology and Biotechnology Letters
    • /
    • v.48 no.3
    • /
    • pp.296-302
    • /
    • 2020
  • Rapamycin, derived from Streptomyces rapamycinicus, is an important bioactive compound having a therapeutic value in managing Parkinson's disease, rheumatoid arthritis, cancer, and AIDS. Because of its pharmaceutical activity, studies over the past decade have focused on the biosynthesis of rapamycin to enhance its yield. In this study, the effect of rapG on rapamycin production was investigated. The rapG expression vector was constructed by utilizing the integration vector pSET152 under the control of the erythromycin resistance gene (ermE), a strong constitutive promoter. The rapamycin yield of wild type (WT) and WT/rapG overexpression mutant strains, under fermentation conditions, was analyzed by high-performance liquid chromatography (HPLC). Our results revealed that overexpression of rapG increased rapamycin production by approximately 4.9-fold (211.4 mg/l) in MD1 containing 15 g/l of glycerol, compared to that of the WT strain. It was also found that Illicium verum powder (10 g/l), containing shikimic acid, enhanced rapamycin production in both WT and WT/rapG strains. Moreover, the amount of rapamycin produced by the WT/rapG strain was statistically higher than that produced by the WT strain. In conclusion, the addition 15 g/l glycerol and 15 g/l I. verum powder produced the optimal conditions for rapamycin production by WT and WT/rapG strains.

Rapamycin reduces orofacial nociceptive responses and microglial p38 mitogen-activated protein kinase phosphorylation in trigeminal nucleus caudalis in mouse orofacial formalin model

  • Yeo, Ji-Hee;Kim, Sol-Ji;Roh, Dae-Hyun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.25 no.4
    • /
    • pp.365-374
    • /
    • 2021
  • The mammalian target of rapamycin (mTOR) plays a role in various cellular phenomena, including autophagy, cell proliferation, and differentiation. Although recent studies have reported its involvement in nociceptive responses in several pain models, whether mTOR is involved in orofacial pain processing is currently unexplored. This study determined whether rapamycin, an mTOR inhibitor, reduces nociceptive responses and the number of Fos-immunoreactive (Fos-ir) cells in the trigeminal nucleus caudalis (TNC) in a mouse orofacial formalin model. We also examined whether the glial cell expression and phosphorylated p38 (p-p38) mitogen-activated protein kinases (MAPKs) in the TNC are affected by rapamycin. Mice were intraperitoneally given rapamycin (0.1, 0.3, or 1.0 mg/kg); then, 30 min after, 5% formalin (10 μl) was subcutaneously injected into the right upper lip. The rubbing responses with the ipsilateral forepaw or hindpaw were counted for 45 min. High-dose rapamycin (1.0 mg/kg) produced significant antinociceptive effects in both the first and second phases of formalin test. The number of Fos-ir cells in the ipsilateral TNC was also reduced by high-dose rapamycin compared with vehicle-treated animals. Furthermore, the number of p-p38-ir cells the in ipsilateral TNC was significantly decreased in animals treated with high-dose rapamycin; p-p38 expression was co-localized in microglia, but not neurons and astrocytes. Therefore, the mTOR inhibitor, rapamycin, reduces orofacial nociception and Fos expression in the TNC, and its antinociceptive action on orofacial pain may be associated with the inhibition of p-p38 MAPK in the microglia.

STRUCTURAL ANALYSIS OF RAPAMYCIN'S ROLE IN BINDING FKBP12 AND FRAP

  • Park, Jungwon;Jie Chen;Stuart Schreiber;Jon Clardy
    • Proceedings of the Korean Biophysical Society Conference
    • /
    • 1996.07a
    • /
    • pp.9-9
    • /
    • 1996
  • The immunosuppressive and cell cycle arrest agent rapamycin works by binding together two proteins: the FK506 binding protein (FKBP12) and the FKBP-rapamycin associated protein (FRAP). A 2.7 $\AA$ resolution crystal structure of the triple complex of human FK506 binding protein (FKBP12), rapamycin, and FKBP12-rapamycin binding domain (FRB) of FRAP, reveals two proteins bound together through rapamycin' s ability to simultaneously occupy two different hydrophobic binding pockets. (omitted)

  • PDF

Rapamycin and PF4 Induce Apoptosis by Upregulating Bax and Down-Regulating Survivin in MNU-Induced Breast Cancer

  • Al-Astani Tengku Din, Tengku Ahmad Damitri;Shamsuddin, Shazana Hilda;Idris, Fauziah Mohamad;Wan Mansor, Wan Nor Ariffin;Abdul Jalal, Muhammad Irfan;Jaafar, Hasnan
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.9
    • /
    • pp.3939-3944
    • /
    • 2014
  • Background: To elucidate the role of rapamycin and PF4 on apoptosis regulation via Bax (pro-apoptosis), Bcl-2 (anti-apoptosis) and survivin activation on the growth in the 1-methyl-1-nitrosourea-induced invasive breast carcinoma model. Materials and Methods: Thirty five female Sprague Dawley rats at age 21-day old were divided into 4 groups; Group 1 (control, n=10), Group 2 (PF4, n=5), Group 3 (rapamycin, n=10) and Group 4 (rapamycin+PF4, n=10). MNU was administered intraperitionally, dosed at 70mg/kg body weight. The rats were treated when the tumors reached the size of $14.5{\pm}0.5mm$ and subsequently sacrificed after 5 days. Rapamycin and PF4 were administered as focal intralesional injections at the dose of $20{\mu}g$/lesion. The tumor tissue was then subjected to histopathological examinations for morphological appraisal and immunohistochemical assessment of the pro-apoptotic marker, Bax and anti-apoptotic markers, Bcl-2 and survivin. Results: The histopathological pattern of the untreated control cohort showed that the severity of the malignancy augments with mammary tumor growth. Tumors developing in untreated groups were more aggressive whilst those in treated groups demonstrated a transformation to a less aggressive subtype. Combined treatment resulted in a significant reduction of tumor size without phenotypic changes. Bax, the pro-apoptotic marker, was significantly expressed at higher levels in the rapamycin-treated and rapamycin+PF4-treated groups compared to controls (p<0.05). Consequently, survivin was also significantly downregulated in the rapamycin-treated and rapamycin+PF4-treated group and this was significantly different when compared to controls (p). Conclusions: In our rat model, it could be clearly shown that rapamycin specifically affects Bax and survivin signaling pathways in activation of apoptosis. We conclude that rapamycin plays a critical role in the induction of apoptosis in MNU-induced mammary carcinoma.

Rapamycin Rescues the Poor Developmental Capacity of Aged Porcine Oocytes

  • Lee, Seung Eun;Kim, Eun Young;Choi, Hyun Yong;Moon, Jeremiah Jiman;Park, Min Jee;Lee, Jun Beom;Jeong, Chang Jin;Park, Se Pill
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.27 no.5
    • /
    • pp.635-647
    • /
    • 2014
  • Unfertilized oocytes age inevitably after ovulation, which limits their fertilizable life span and embryonic development. Rapamycin affects mammalian target of rapamycin (mTOR) expression and cytoskeleton reorganization during oocyte meiotic maturation. The goal of this study was to examine the effects of rapamycin treatment on aged porcine oocytes and their in vitro development. Rapamycin treatment of aged oocytes for 24 h (68 h in vitro maturation [IVM]; $44h+10{\mu}M$ rapamycin/24 h, $47.52{\pm}5.68$) or control oocytes (44 h IVM; $42.14{\pm}4.40$) significantly increased the development rate and total cell number compared with untreated aged oocytes (68 h IVM, $22.04{\pm}5.68$) (p<0.05). Rapamycin treatment of aged IVM oocytes for 24 h also rescued aberrant spindle organization and chromosomal misalignment, blocked the decrease in the level of phosphorylated-p44/42 mitogen-activated protein kinase (MAPK), and increased the mRNA expression of cytoplasmic maturation factor genes (MOS, BMP15, GDF9, and CCNB1) compared with untreated, 24 h-aged IVM oocytes (p<0.05). Furthermore, rapamycin treatment of aged oocytes decreased reactive oxygen species (ROS) activity and DNA fragmentation (p<0.05), and downregulated the mRNA expression of mTOR compared with control or untreated aged oocytes. By contrast, rapamycin treatment of aged oocytes increased mitochondrial localization (p<0.05) and upregulated the mRNA expression of autophagy (BECN1, ATG7, MAP1LC3B, ATG12, GABARAP, and GABARAPL1), anti-apoptosis (BCL2L1 and BIRC5; p<0.05), and development (NANOG and SOX2; p<0.05) genes, but it did not affect the mRNA expression of pro-apoptosis genes (FAS and CASP3) compared with the control. This study demonstrates that rapamycin treatment can rescue the poor developmental capacity of aged porcine oocytes.