手机版
帕金森病 Parkinson's disease
帕站首页 专题讨论 帕金森病 放心医生 中医与帕 病友故事 留言交流 脑起搏器 专家解答
 
第 1 2 3 4 5 6 7 8 9 10 31 页 (第2页,共306条)
薛传恽:回复9楼 异乡人  邮箱:xxuuee@gmail.com  IP:203.186.64.167  日期:2013-9-9 [回复11楼]

  回复9楼 异乡人
  回复9楼 异乡人:回复8楼 Andy
  
  哈哈!我真的这次没有听明白。因为我现在已经习惯于用广东话来看书念字了。在广东话里克字念赫或盒。三盒油就无论如何联想不到Thank you了。
  Andy是鸡我是鸭,便互相听不懂了。
   

薛传恽:回复1楼 薛传恽  邮箱:xxuuee@gmail.com  IP:210.6.141.246  日期:2013-9-9 [回复12楼]

  回复1楼 薛传恽
  回复1楼 薛传恽:令人兴奋的新消息
  
  看了许多跟贴,给我一个感觉,似乎还没有谁愿意做鲁迅说的第一个吃螃蟹的人。
  其实现在吃已不是第一个吃螃蟹了。可能上万的人已经先吃了。可靠性和副作用已是可以接受的了。和医生谈谈试吃吧!可以推迟甚至停止病情发展。效果应该超过雷沙吉兰。 

余军:回复12楼 薛传恽  IP:27.224.160.170  日期:2013-9-9 [回复13楼]

  回复12楼 薛传恽
  回复12楼 薛传恽:回复1楼 薛传恽
  
  谢谢薛老给我们带来的振奋人心的消息,我明天就去买来吃,到时候再回来向大家汇报。 

异乡人:回复11楼 薛传恽  IP:114.222.237.185  日期:2013-9-9 [回复14楼]

  回复11楼 薛传恽
  回复11楼 薛传恽:回复9楼 异乡人
  
  
  熊去氧胆酸片
  【生产厂家】上海信谊药厂有限公司
  
  【产品规格】50mg*30s
  
  【产品包装】400
  
  【计价单位】瓶
  
  【销售价格】¥7.68
  
   

异乡人:回复11楼 薛传恽  IP:114.222.237.185  日期:2013-9-9 [回复15楼]

  回复11楼 薛传恽
  回复11楼 薛传恽:回复9楼 异乡人
  
  薛老不仅给患者带来抗帕的希望,也经常给网站带来轻松的气氛。谢谢啦! 

余军:回复12楼 薛传恽  IP:27.224.160.170  日期:2013-9-9 [回复16楼]

  回复12楼 薛传恽
  回复12楼 薛传恽:回复1楼 薛传恽
  
  薛老您好,感谢您不辞辛苦多年来为帕金森病友所做出的努力。能不能再辛苦您把那篇文章的全部内容搬来给我们帕友们分享一下呢,因为这个幸福来得太突然了,但愿不是空欢喜一场。 

薛传恽:回复16楼 余军  邮箱:xxuuee@gmail.com  IP:210.6.141.246  日期:2013-9-10 [回复17楼]

  回复16楼 余军
  回复16楼 余军:回复12楼 薛传恽
  
  全文搬出来了。原文的格式很复杂,在这里表现不出来。被这个网站约束以后的样子就如下所示。一些图表都不见了。不过没关系,大家只要看个大概就够。我要翻译不照他。不过翻译会很艰苦。我现在效率很低。大家要多等些时间。
  
  Ursocholanic acid rescues mitochondrial function
  in common forms of familial Parkinson’s disease
  Heather Mortiboys,1 Jan Aasly2 and Oliver Bandmann1
  1 Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
  2 Department of Neurology, St Olav’s Hospital, Trondheim, Norway
  Correspondence to: Oliver Bandmann, MD PhD
  Sheffield Institute for Translational Neuroscience (SITraN),
  Department of Neuroscience,
  University of Sheffield,
  385a Glossop Road,
  Sheffield S10 2HQ, UK
  E-mail: o.bandmann@sheffield.ac.uk
  Previous drug screens aiming to identify disease-modifying compounds for Parkinson’s disease have typically been based on
  toxin-induced in vitro and in vivo models of this neurodegenerative condition. All these compounds have failed to have a reliable
  disease-modifying effect in subsequent clinical trials. We have now established a novel approach, namely to screen an entire
  compound library directly in patient tissue to identify compounds with a rescue effect on mitochondrial dysfunction as a crucial
  pathogenic mechanism in Parkinson’s disease. The chosen Microsource Compound library contains 2000 compounds, including
  1040 licensed drugs and 580 naturally occurring compounds. All 2000 compounds were tested in a step-wise approach for their
  rescue effect on mitochondrial dysfunction in parkin (PARK2) mutant fibroblasts. Of 2000 compounds, 60 improved the mitochondrial
  membrane potential by at least two standard deviations. Subsequently, these 60 compounds were assessed for their toxicity
  and drug-like dose-response. The remaining 49 compounds were tested in a secondary screen for their rescue effect on intracellular
  ATP levels. Of 49 compounds, 29 normalized ATP levels and displayed drug-like dose response curves. The mitochondrial rescue
  effect was confirmed for 15 of these 29 compounds in parkin-mutant fibroblasts from additional patients not included in the initial
  screen. Of 15 compounds, two were chosen for subsequent functional studies, namely ursocholanic acid and the related compound
  dehydro(11,12)ursolic acid lactone. Both compounds markedly increased the activity of all four complexes of the mitochondrial
  respiratory chain. The naturally occurring compound ursolic acid and the licensed drug ursodeoxycholic acid are chemically closely
  related to ursocholanic acid and dehydro(11,12)ursolic acid lactone. All four substances rescue mitochondrial function to a similar
  extent in parkin-mutant fibroblasts, suggesting a class effect. The mitochondrial rescue effect depends on activation of the
  glucocorticoid receptor with increased phosphorylation of Akt and was confirmed for both ursocholanic acid and ursodeoxycholic
  acid in a Parkin-deficient neuronal model system. Of note, both ursocholanic acid and ursodeoxycholic acid also rescued mitochondrial
  function in LRRK2G2019S mutant fibroblasts. Our study demonstrates the feasibility of undertaking drug screens in
  Parkinson’s disease patients’ tissue and has identified a group of chemically-related compounds with marked mitochondrial
  rescue effect. Drug repositioning is considered to be a time- and cost-saving strategy to assess drugs already licensed for a different
  condition for their neuroprotective effect. We therefore propose both ursolic acid as a naturally occurring compound, and ursodeoxycholic
  acid as an already licensed drug as promising compounds for future neuroprotective trials in Parkinson’s disease.
  Keywords: Parkinson’s disease; parkin; LRRK2; mitochondria; disease-modifying therapy
  Abbreviations: DUA = dehydro(11,12)ursolic acid lactone; MPTP = 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
  doi:10.1093/brain/awt224 Brain 2013: Page 1 of 13 | 1
  Received January 21, 2013. Revised May 29, 2013. Accepted June 9, 2013.
   The Author (2013). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved.
  For Permissions, please email: journals.permissions@oup.com
  Brain Advance Access published September 2, 2013
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  Introduction
  Parkinson’s disease is a common and relentlessly progressive,
  incurable neurodegenerative condition. Its world-wide prevalence
  is expected to double by 2030 (Dorsey et al., 2007). Currently
  available drugs only result in symptomatic improvement with
  limited efficacy. In the past, compounds were typically tested for
  their putative neuroprotective effect in toxin-induced, in vitro and
  in vivo models of Parkinson’s disease. However, exposure to
  toxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
  (MPTP) only partially resembles the mechanisms leading to
  Parkinson’s disease, if at all. Subsequently undertaken clinical
  trials failed to confirm a beneficial, disease-modifying effect for
  any compound with a promising initial effect in these traditional
  MPTP models (Lang, 2006). Mitochondrial dysfunction is a key
  mechanism in the pathogenesis of both sporadic and familial
  Parkinson’s disease (Exner et al., 2012). Mutations in the autosomal
  recessively inherited parkin (also known as PARK2) gene are
  the most common identifiable cause of early-onset Parkinson’s
  disease. The LRRK2G2019S mutation is the most common identifiable
  cause of monogenically inherited late-onset Parkinson’s disease
  (Hardy, 2010). We have previously demonstrated abnormal
  mitochondrial function with specific lowering of complex I activity
  of the mitochondrial respiratory chain in skin fibroblasts of parkinmutant
  patients with Parkinson’s disease (Mortiboys et al., 2008).
  We and others subsequently also reported mitochondrial dysfunction
  in fibroblasts from patients with the LRRK2G2019S mutation
  (Mortiboys et al., 2010; Papkovskaia et al., 2012).
  The aim of this study was to undertake an in vitro compound
  screen in Parkinson’s disease mutant patient tissue to identify
  mitochondrial rescue compounds. Our project is based on the hypothesis
  that any compound with a robust mitochondrial rescue
  effect in Parkinson’s disease patient tissue is more likely to exert a
  subsequent beneficial effect in clinical trials than those compounds
  that have only been tested in toxin-induced model systems. Two
  thousand compounds from the Microsource Spectrum Collection
  (www.msdiscovery.com) were assessed for their rescue effect on
  mitochondrial function in several stages. This compound library
  consists of 1040 licensed drugs, 580 natural compounds and
  420 other bioactive compounds. The large proportion of licensed
  drugs and natural compounds made it plausible to assume that
  any positive hits in our compound screen could rapidly be taken
  into clinical trials.
  Materials and methods
  Patients
  The project was reviewed by the local ethics committee. Informed
  consent was taken from all research participants (see Supplementary
  Table 1 for further information on all patients included in this study).
  There was no significant difference in age between the four parkinmutant
  patients and their four matched controls (age in years  SD
  parkin-mutant patients, 40.5  6.5; controls, 38.5  5.5). Similarly,
  there was no significant difference in age between the three
  LRRK2G2019S mutant patients and their three matched controls
  (LRRK2G2019S mutant patients, age 59  5.5; controls, age 61  4.5).
  Groups were also sex matched.
  Methods
  Fibroblast cell culture conditions as well as measurement of mitochondrial
  membrane potential, respiratory chain function and cellular ATP
  production were carried out as previously described (Mortiboys et al.,
  2008).
  Z-scores
  In order to assess the robustness and reproducibility of the assays used
  as primary and secondary screens we undertook rigorous testing using
  Z’ and SW score calculations as described (http://www.ncats.nih.
  gov/). See Supplementary material for further information.
  Primary drug screen
  Stage 1
  Parkin-mutant fibroblasts from two parkin-mutant patients were incubated
  with all 2000 Microsource Spectrum Collection compounds for
  24 h at a concentration of 10 mM. Each drug treatment was carried out
  in duplicate, thus, a total of four drug exposure experiments were
  carried out at the first stage for each compound. A positive hit was
  defined a priori as a compound that would improve the mitochondrial
  membrane potential by more than 3 standard deviations (SD) in at
  least three of the experiments and by at least 2 SD in the fourth
  experiment. Positive hits were then tested further in cell-free assays
  to exclude a possible false-positive effect due to autofluorescence of
  the drug or a drug interaction with tetramethylrhodamine methyl ester
  (TMRM). In addition, compounds were tested for any cellular toxicity
  effects using the lactate dehyodrogenase (LDH) assay as described
  previously (Mortiboys et al., 2008). Furthermore, dose-response
  assessments (0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30 and 100 mM) were
  undertaken to determine the shape of the dose response curves.
  Stage 2
  Positive hits from the Stage 1 experiments were assessed for their
  rescue effect on intracellular ATP levels. As before, parkin-mutant
  fibroblasts from the same two patients and matched controls were
  treated twice at a concentration of 10 mM for 24 h. A positive hit at
  Stage 2 was again defined as a compound that improved intracellular
  ATP levels by at least 3 SD in at least three experiments and by at least
  2 SD in the fourth. Positive hits were tested for dose response curves
  (0.01–100 mM) again. Positive hits with a sigmoidal dose-response
  curve were tested for their recovery effects on ATP levels in an additional
  two parkin-mutant patient and matched control fibroblast lines.
  Stage 3
  Selected top hits from Stage 2 were then assessed further for their
  effect on the four individual mitochondrial respiratory chain complexes
  in fibroblasts from four parkin-mutant patients and matched control
  subjects. Fibroblasts (1.4  107 cells) were treated for 24 h with
  100nM of each compound before being harvested by trypsinization
  and used for all further analyses. Mitochondrially enriched fractions
  and individual mitochondrial respiratory chain assays were all done
  as described previously (Mortiboys et al., 2008). All data are expressed
  to mg protein. Protein was measured using the Bradford assay (Peirce)
  as per the manufacturers’ instructions.
  2 | Brain 2013: Page 2 of 13 H. Mortiboys et al.
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  Functional studies
  Pharmacological inhibition of glucocorticoid receptor
  Fibroblasts were plated (5000 cells per well) into 96 well plates. After
  24 h, cells were treated with 1 mM RU486 for 4 h before adding either
  100nM of selected compounds (see ‘Results’ section). Cellular ATP
  levels were measured 24 h later as described above.
  Small interfering RNA glucocorticoid receptor
  knockdown
  Small interfering RNA oligonucleotides were targeted to the
  glucocorticoid receptor gene (NR3C1), target sequence AAGTG
  CAAACCTGCTGTGTTT or scramble control small interfering RNA
  (both Qiagen). Small interfering RNAs (10 nM) (NC3C1-targeted or
  scramble negative) were transfected into fibroblasts using 0.5mM
  Lipofectamine 2000 according to the manufacturers’ instructions.
  Knockdown efficiency of the glucocorticoid receptor protein was
  assessed using the glucocorticoid receptor ELISA (Abnova) at 48 h
  post-transfection as per the manufacturer’s instructions. Twenty-four
  hours post-transfection cells were treated with 100nM of selected
  compounds; cellular ATP levels were measured 24 h later as described
  above.
  Quantification of total Akt and phosphorylated
  Akt at Ser473
  Akt and phosphorylated (p)Akt Ser473 ELISAs (Invitrogen) were performed
  on fibroblast cell lysates as per the manufacturer’s instructions
  using the provided standards to calculate the amount of protein present.
  All data are presented as a ratio of pAkt (Ser473): total Akt.
  Pharmacological inhibition of the Akt pathway
  Fibroblasts were plated (5000 cells per well) into 96 well plates. After
  24 h, cells were treated with 1 mM LY294002 or 50nM triciribine for
  15 min before adding 100nM of selected compounds. Cellular ATP
  levels were measured 24 h later as described above.
  Confirmatory experiments
  Mouse cortical neurons were prepared from embryonic Day 15 mouse
  embryos as previously described (Kasher et al., 2009). Approximately
  6  104 neurons were plated into each well of a 96-well plate (previously
  coated with poly-L-lysine) or 2  105 neurons were plated into
  each well of a 24-well plate for either ATP assays or harvesting for
  western blot analysis or fixed for imaging. After 5 days in culture
  neurons we transfected using the Accell siRNA and Accell siRNA
  media (as per the manufacturer’s instructions, Dharmacon) with
  either scramble-negative control small interfering RNA (Accell mouse
  control siRNA kit, Dharmacon) or parkin small interfering RNA
  (sequence GUUUCCACUUGUAUUGUGU). Forty-eight hours posttransfection
  neurons were dosed with various concentrations of compounds
  and 24 h later the cellular ATP assay was performed as
  described above, or neurons were harvested for western blotting.
  Western blotting was performed as described previously (Mortiboys
  et al., 2008). Coverslips were fixed with 4% paraformaldehyde for
  30 min with subsequent PBS washes. Cells were permeabilized with
  0.1% TritonTM X-100 for 10 min at room temperature and blocked
  with 1% goat serum for 1 h. Cells were incubated with primary antibodies
  (rabbit anti-Parkin; Abcam and mouse anti-TOM-20; BD
  Biosciences) at 1:500 overnight at 4C with subsequent PBS washes
  and incubation with rabbit anti-mouse and goat anti-rabbit secondary
  antibodies for 1 h at room temperature. Cells were stained with
  Hoescht and then mounted into glass slides using ProLong Gold
  (Invitrogen).
  Cellular ATP levels were measured as described previously
  (Mortiboys et al., 2008) in fibroblasts from three LRRK2G2019S
  mutant patients with Parkinson’s disease and three age and sexmatched
  controls. The cells were treated with 100nM of the selected
  compounds for 24 h before measurement.
  Statistical analysis
  Values from multiple experiments were expressed as means  SE (standard
  error). Statistical significance (Bonferroni corrected) was assessed
  using Student’s t-test for data with a normal distribution, a non-parametric
  t-test was used for data with a skewed distribution. The effect of
  multiple factors was assessed using a two-way ANOVA test.
  Results
  A summary of our screening strategy is given in Fig. 1. Of 2000
  compounds, 60 improved the mitochondrial membrane potential
  in parkin-mutant fibroblasts by 43 SD in three of the four
  experiments and by 42 SD in the fourth. Two compounds elicited
  an increase in the TMRM fluorescence signal in subsequent cellfree
  assays and were thus excluded as false-positive. A further
  nine compounds had to be excluded due to their toxicity
  (Table 1). Full dose-response curves were established for all 49
  remaining compounds, which were then also further assessed for
  their effect on total intracellular ATP levels. Of 49 compounds, 35
  increased the ATP levels in the parkin-mutant fibroblasts by 43
  SD in at least three experiments and by 42 SD in the fourth
  (Table 1). Full dose-response curves were carried out using these
  top 35 compounds. Six compounds did not display a drug-like,
  sigmoidal dose response curve and were therefore excluded, leaving
  29 compounds.
  Each of these 29 compounds was then tested for their rescue
  effect on cellular ATP levels in a further two patient fibroblast lines
  and two control fibroblast lines. Of 29 compounds, 15 rescued
  cellular ATP levels by 43 SD in all four parkin-mutant fibroblast
  lines tested (Table 2).
  Of 15 compounds, two, namely ursocholanic acid and dehydro
  (11,12) ursolic acid lactone (DUA), were selected for further
  assessment. Reasons for not investigating the remaining 13 compounds
  forward at this stage are listed in Table 2 and, in greater
  detail, in the Supplementary material. Ursocholanic acid and DUA
  were then further assessed for their effect on the activity of complexes
  I–IV of the respiratory chain. Ursocholanic acid significantly
  rescued and increased the activity of complexes I–IV by 200–
  500% (Fig. 2). Treatment with DUA achieved very similar results
  (Supplementary Fig. 1).
  Interestingly, 7 of the 15 Stage 2 positive hits were steroids or
  related compounds with four carbon rings forming the (steroid)
  backbone of each particular compound, including ursocholanic
  acid and DUA (Table 2). We therefore hypothesized that their
  observed rescue effect was mediated through activation of the
  glucocorticoid receptor. To further test this hypothesis, parkinmutant
  cells were pretreated with the glucocorticoid receptor antagonist
  RU486 to determine whether glucocorticoid receptor
  Ursocholanic acid in familial PD Brain 2013: Page 3 of 13 | 3
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  inhibition may abolish the observed rescue effect of DUA and
  ursocholanic acid on ATP levels. As predicted, RU486 completely
  eliminated the rescue effect of all tested compounds on cellular
  ATP levels (Fig. 3A). To further validate these results, we used a
  different method, namely small interfering RNA-mediated glucocorticoid
  receptor knockdown before treatment with ursocholanic
  acid or DUA. Glucocorticoid receptor protein knockdown was confirmed
  to be 75%  3.8% (mean  SD) using ELISA at 48 h post
  transfection (data not shown). As predicted, small interfering
  RNA-mediated glucocorticoid receptor knockdown abolished the
  rescue effect of 100nM ursocholanic acid or DUA on intracellular
  ATP levels (Fig. 3B).
  Ursolic acid and ursodeoxycholic acid were not part of the initially
  screened Microsource Compound Library, but are chemically related
  to ursocholanic acid and DUA (Fig. 4). Ursolic acid exerts its beneficial
  effect on muscle atrophy through Akt activation, namely by
  increased phosphorylation of Akt at Ser473 (Kunkel et al., 2011).
  Similarly, ursodeoxycholic acid exerts its protective effect against
  mitochondria-dependent programmed cell death in SH-SY5Y cells
  through Akt activation (Chun and Low, 2012).
  We therefore assessed the effect of DUA and ursocholanic acid
  in parkin-mutant fibroblasts on Akt phosphorylation at Ser473.
  There was a marked increase in the pAktSer473:Akt protein ratio
  by 400% after treatment with DUA and 305% after treatment
  with ursocholanic acid (P50.05) in parkin-mutant fibroblasts
  compared with the ratio in untreated parkin-mutant fibroblasts
  (Fig. 5A). Interestingly, this change was only evident in parkinmutant
  fibroblasts, the pAktSer473:Akt ratio in control fibroblasts
  remained constant after drug treatment. We next aimed to confirm
  that both ursocholanic acid and DUA are exerting their mitochondrial
  rescue effect through activation of the Akt pathway
  rather than Akt activation merely being associated with the
  rescue effect of our top compounds. As predicted, pretreatment
  with either Akt inhibitor LY29400 (a phosphatidylinositol 3-kinase
  inhibitor) or triciribine (a selective inhibitor of cellular phosphorylation/
  activation of Akt) abolished the rescue effect of ursocholanic
  acid and DUA on cellular ATP levels in parkin-mutant fibroblasts
  (Fig. 5B).
  Neither DUA nor ursocholanic acid are FDA-licensed drugs; little
  information is available on their bioavailability and safety in humans.
  In contrast, the chemically closely related bile acid ursodeoxycholic
  acid has been in clinical use as treatment for primary biliary cirrhosis
  for 430 years. Its clinical pharmacokinetics are well characterized
  (Ward et al., 1984). The chemically closely related ursolic acid is a
  naturally occurring compound present in many plants. Based on their
  structural similarities, we hypothesized that both ursolic acid and
  ursodeoxycholic acid may have a similar mitochondrial rescue
  effect as DUA and ursocholanic acid. Indeed, both ursolic acid and
  ursodeoxycholic acid normalized intracellular ATP levels similar to the
  effect observed for DUA and ursocholanic acid (Fig. 6).
  Effect in Parkin-deficient neuronal
  model system
  We next assessed the rescue effect of ursocholanic acid and ursodeoxycholic
  acid in a neuronal cell culture model. Small interfering
  RNA mediated knockdown of parkin resulted in a reduction of Parkin
  protein levels by 80% in cortical mouse neurons as shown by western
  blotting and a decrease in cellular ATP levels by 40%. Treatment with
  10pM ursocholanic acid or 10pM ursodeoxycholic acid rescued the
  cellular ATP loss in these Parkin-deficient neurons (Fig. 7). Thus,
  ursocholanic acid and ursodeoxycholic acid have a rescue effect on
  mitochondrial dysfunction not only in parkin-mutant fibroblasts but
  also in parkin-deficient neurons.
  Rescue effect in LRRK2G2019S mutant
  patient tissue
  We finally determined whether ursocholanic acid and the chemically
  related and FDA-licensed drug ursodeoxycholic acid also
  have a mitochondrial rescue effect in other forms of familial
  Figure 1 This flowchart shows an overview of the screening strategy used. Each part of the screen is depicted as is the number of positive
  hit compounds that were taken to the next stage of the screen.
  4 | Brain 2013: Page 4 of 13 H. Mortiboys et al.
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  Table 1 Positive hits of the primary screen, and results from stage 1 and 2 of the drug screen
  Drug name Stage 1 Stage 2
  Cell
  free
  Toxicity EC50
  MMP
  ATP
  recovery
  Dose
  response
  curve
  EC50
  ATP
  Podophyllotoxin acetate 3 3 Ambiguous X X X
  2,6-Dimethoxyquinone 3 3 1 mM X X X
  Ginkgolic acid 3 3 1.6mM 3 3 250nM
  2’,Beta-dihydroxychalcone 3 3 200nM 3 3 250nM
  Gatifloxacin 3 3 100nM 3 3 250nM
  Amlodipine besylate 3 3 1 mM 3 3 250nM
  Simvastatin 3 3 1 mM X X X
  Hydroquinone 3 3 1 mM X X X
  7-Methoxychromone 3 3 100nM X X X
  Perindopril erbumine 3 3 12 mM 3 3 150nM
  Ceftibuten 3 3 1 mM 3 3 250nM
  Cefdinir 3 3 25 mM 3 3 350nM
  3Alpha-hydroxy-3-deoxyangolensic acid methyl ester 3 3 100nM 3 3 150nM
  Dibenzothiophene 3 3 600nM X X X
  Clonidine hydrochloride 3 3 1 mM X X X
  Desipramine hydrochloride 3 X X X X X
  Ginkgolide a 3 3 100nM 3 3 150nM
  Sericetin 3 3 158nM 3 3 150nM
  Friedelin 3 3 1 mM 3 3 150nM
  3Beta,7beta-diacetoxydeoxodeacetoxydeoxydihydrogedunin 3 3 240nM X X X
  Oleanolic acid acetate 3 X X X X X
  Pristimerol diacetate 3 3 631 mM 3 3 125nM
  Khellin 3 3 6 mM 3 3 250nM
  Khivorin 3 3 6 mM X X X
  Allopurinol 3 3 1 mM X X X
  Menthone 3 X 7mM X X X
  Acetylcholine 3 X 60 mM X X X
  Probenecid 3 X 13 mM X X X
  Enalapril maleate 3 X 2mM X X X
  Acivicin 3 X 31mM X X X
  Ephedrine (1 R,2S) hydrochloride 3 3 Ambiguous 3 X X
  Propylthiouracil 3 3 Ambiguous 3 X X
  Clobetasol propionate 3 3 10 mM 3 3 1 mM
  Santonin 3 3 125nM X X X
  Ursocholanic acid 3 3 1 mM 3 3 350nM
  Methylergonovine maleate 3 3 Ambiguous 3 X X
  Androsterone sodium sulfate 3 3 5mM 3 3 350nM
  Dehydro (11,12)ursolic acid lactone (no longer available) 3 3 100 mM 3 3 350nM
  Cholest-5-en-3-one 3 3 1 mM 3 X X
  Fluorometholone 3 3 350nM 3 X X
  Prazosin hydrochloride 3 3 250nM 3 3 150nM
  Narasin 3 X X X X X
  Cedryl acetate 3 X X X X X
  N-benzyltropan-4-ol X 3 X X X X
  Naproxol X 3 X X X X
  Hydroxychloroquine sulphate 3 3 1.2mM 3 3 1 mM
  11-Oxoursolic acid acetate (no longer available) 3 3 0.1nM 3 3 150nM
  Prednisolone 3 3 446mM 3 3 350nM
  Ebselen 3 3 100nM 3 3 1 mM
  Racephedrine hydrochloride 3 3 5 mM X X X
  Snap (S-nitroso-N-acetylpenicillamine) 3 3 200nM X X X
  3-Amino-beta-pinene 3 3 10 mM 3 3 1 mM
  Benzalkonium chloride 3 3 12.5nM 3 3 1 mM
  Melezitose 3 3 1 mM 3 3 1 mM
  (continued)
  Ursocholanic acid in familial PD Brain 2013: Page 5 of 13 | 5
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  Parkinson’s disease. We therefore investigated the effect of these
  compounds on cellular ATP levels in LRRK2G2019S mutant patient
  tissue. Treatment of LRRK2G2019S mutant fibroblasts from three
  different patients with Parkinson’s disease carrying this mutation
  with 10nM of ursocholanic acid or ursodeoxycholic acid for 24 h
  resulted in complete rescue of cellular ATP levels (Fig. 8), similar to
  the effect observed in parkin-mutant patient tissue. Therefore, the
  beneficial effect of these compounds does not appear to be limited
  to parkin-associated Parkinson’s disease.
  Discussion
  The strong evidence of mitochondrial dysfunction in both sporadic
  and familial Parkinson’s disease suggests targeting mitochondria as
  a promising strategy for disease-modifying therapy in Parkinson’s
  disease (Meissner et al., 2011; Schapira, 2012). We had previously
  demonstrated a complete rescue of mitochondrial dysfunction in
  parkin-mutant patient tissue using the glutathione precursor
  Table 1 Continued
  Drug name Stage 1 Stage 2
  Cell
  free
  Toxicity EC50
  MMP
  ATP
  recovery
  Dose
  response
  curve
  EC50
  ATP
  3-Oxoursan (28-13)olide 3 3 1 mM X X X
  Budesonide 3 3 390nM 3 3 1 mM
  Prednisolone acetate 3 3 150nM 3 3 1 mM
  Furegrelate sodium 3 3 3.9nM X X X
  Tamoxifen citrate 3 3 1nM 3 3 1 mM
  6,7-Dichloro-3-hydroxy-2-quinoxalinecarboxylic acid 3 3 Ambiguous 3 X X
  This table details the positive hits of the primary screen and the results from each part of stage 1 and stage 2 of the drug screen. 3 indicates that the compound fulfilled the
  necessary criteria and went through this particular stage; X indicates it did not and was therefore not taken any further. ‘Cell free’ indicates whether the compound reacted
  with the fluorescent dye tetramethylrhodamine methyl ester (TMRM) in a cell free assay. The ‘Tox’ column provides information on possible toxicity of the respective
  compound. ‘EC50 MMP’ indicates the EC50 concentration of the compounds in the mitochondrial membrane potential assay. ‘ATP recovery’ indicates if the compounds
  were also effective in recovering the ATP levels in parkin-mutant fibroblasts. ‘Dose response curve’ indicates whether the compounds displayed a known characterised dose
  response curve shape. ‘EC50 ATP’ provides information about the EC50 of the compounds in the cellular ATP assay.
  Table 2 Top 15 hits that rescued the mitochondrial membrane potential and cellular ATP levels in all four patients and had
  drug-like dose response curves
  Drug name Compound origin Steroid like
  structure
  Additional comments
  Gatifloxacin Synthetic X Antibiotic with negative effect on glucose homeostasis and
  neurological function in vivo
  Amlodipine besylate Synthetic X Ca-antagonist, concerns about side-effect profile (including
  oedema, insomnia, dizziness, depression)
  3Alpha-hydroxy-3-deoxyangolensic
  acid methyl ester
  Natural X No information on use in humans or rodents
  Ginkgolide a Natural X Previous studies have given inconsistent results for
  neuroprotective effect of ginkgo in neurodegenerative
  disease and related model systems
  Pristimerol diacetate Semi synthetic X No information on use in humans or rodents
  Ephedrine (1R,2S) hydrochloride Natural X Sympatomimetic amine, intolerance and drug interaction
  likely in Parkinson’s disease
  Ursocholanic acid Natural 3 Taken forward
  Androsterone sodium sulphate Semi synthetic 3 Steroid, excluded due to likelihood of side effects on long
  term treatment
  Dehydro (11,12)ursolic acid lactone Natural 3 Taken forward
  Cholest-5-en-3-one Semi synthetic 3 cholesterol, excluded due to likelihood of side effects on
  long term treatment
  Hydroxychloroquine sulphate Synthetic X Inhibitory effect on mitophagy
  11-Oxoursolic acid acetate Natural 3 Unable to obtain more of the compound
  Budesonide Semi synthetic 3 Steroid with high-first pass effect, excluded due to likelihood
  of limited biological availability
  Prednisolone acetate Semi synthetic 3 Steroid, excluded due to likelihood of side effects on long
  term treatment
  Tamoxifen citrate Synthetic X Can cause cognitive impairment and other major side effects
  Additional information is provided on origin of compound, the presence of a steroid-like structure as well as justification for not taking the majority of these compounds
  forward. The two compounds taken forward are the chemically related substances ursocholanic acid and dehydro (11,12) ursolic acid lactone. Additional information on
  those compounds that have been excluded from further analysis is provided in the Supplementary material.
  6 | Brain 2013: Page 6 of 13 H. Mortiboys et al.
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  L-2-oxothiazolidine-4-carboxylic acid (OTCA) and also a mild partial
  rescue effect on mitochondrial function after rapamycin treatment
  (Mortiboys et al., 2008; Tain et al., 2009). Based on these
  ‘proof of principle’ data, we have now undertaken the first drug
  screen in Parkinson’s disease patient tissue and identified a group
  of chemically-related compounds with marked rescue effect on
  mitochondrial function. Our data are in keeping with previous
  studies that reported a protective effect of the taurine conjugate
  of ursodeoxycholic acid (TUDCA) against mitochondrial toxins in
  parkin-deficient Caenorhabditis elegans (Ved et al., 2005).
  Recently, an Akt-mediated, partial neuroprotective effect of
  TUDCA on MPTP-induced dopaminergic cell death has been
  observed in a mouse model of Parkinson’s disease (Castro-
  Caldas et al., 2012). Our data strongly suggest a class effect for
  bile acids and their derivates such as DUA, ursocholanic acid and
  ursodeoxycholic acid and the natural pentacyclic triterpenoid
  Figure 2 Rescue of mitochondrial function in parkin-mutant fibroblasts by treatment with 100nM ursocholanic acid (UCA) for 24 h.
  (A) Mitochondrial membrane potential and (B) cellular ATP levels are decreased in untreated fibroblasts of patients with parkin mutations
  compared with untreated controls (P50.05), treatment with ursocholanic acid results in normalization of mitochondrial membrane
  potential and ATP levels (P50.05). (C) Mitochondrial membrane potential and (D) cellular ATP levels after treatment with increasing
  concentrations of ursocholanic acid for 24 h, reflecting a sigmoidal dose response curve. (E and F) Activity of each of the individual
  respiratory chain enzymes are increased by treatment with ursocholanic acid in both control and parkin-mutant fibroblasts. Data presented
  are corrected to protein levels *P50.05, **P50.01, ***P50.001.
  Ursocholanic acid in familial PD Brain 2013: Page 7 of 13 | 7
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  ursolic acid. The bioavailability of ursolic acid and its dose-dependent
  increase in brain tissue of mice has been well characterized
  (Yin et al., 2012). A beneficial effect of both ursolic acid and
  ursodeoxycholic acid or TUDCA has also been described in different
  in vitro and in vivo model systems for other
  neurodegenerative conditions, including Alzheimer’s disease,
  Huntington’s disease and stroke (Keene et al., 2002; Rodrigues
  et al., 2003; Ramalho et al., 2008; Wilkinson et al., 2011).
  Of note, 7 of 15 of the compounds that rescued both the mitochondrial
  membrane potential and cellular ATP levels as well as
  Figure 3 Inhibition or knockdown of the glucocorticoid receptor abolishes the rescue effect of ursocholanic acid (UCA) and DUA in
  parkin-mutant fibroblasts. (A) Cellular ATP levels are reduced in parkin-mutant patient fibroblasts (black bars) compared with controls
  (white bars) and recovered to normal levels after treatment with 100nM ursocholanic acid or DUA for 24 h. This rescue effect is completely
  abolished by pretreatment with 1 mM RU486 (glucocorticoid receptor antagonist) for 4 h. (B) Cellular ATP levels are reduced in
  parkin mutant patient fibroblasts transfected with either scramble small interfering RNA (dark grey bars) or glucocorticoid receptor small
  interfering RNA (black bars) compared with control fibroblasts transfected with scramble small interfering RNA (white bars) or glucocorticoid
  receptor small interfering RNA (light grey bars) *P50.05. Treatment with 100nM ursocholanic acid or DUA completely rescues
  this defect in parkin mutant fibroblasts transfected with scramble small interfering RNA (white and dark grey bars) but not in parkin
  mutant fibroblasts transfected with glucocorticoid receptor small interfering RNA treatment with ursocholanic acid and DUA (black bars)
  compared with controls also transfected with glucocorticoid receptor small interfering RNA (light grey bars). DMSO = dimethylsulphoxide.
  Figure 4 Structures of the top two compounds identified from the original drug screen, namely (A) dehydro (11,12) ursolic acid lactone and
  (C) ursocholanic acid and two further compounds which are structurally similar, namely (B) ursolic acid and (D) ursodeoxycholic acid. The
  structural similarities are highlighted in red. The structures are represented in standard chemical format displaying the 3D orientation of
  groups. Where no group is specified a methyl group is attached. Hydrogens are only shown if they affect the 3D orientation of the molecule.
  8 | Brain 2013: Page 8 of 13 H. Mortiboys et al.
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  having drug-like dose response curves had a steroid-like structure.
  Lim et al. (2012) reported independently a neuroprotective effect
  of the chemically closely related sterol biosynthesis intermediate
  lanosterol. Both ursolic acid and lanosterol induce mild mitochondrial
  uncoupling that has been proposed as a promising strategy
  for disease modification in Parkinson’s disease (Liobikas et al.,
  2011; Ho et al., 2012; Lim et al., 2012).
  The inhibition of the mitochondrial rescue effect of DUA and
  ursocholanic acid after pretreatment with RU486 is in keeping
  with previous observations on glucocorticoid receptor-mediated
  biological activity of ursolic acid or ursodeoxycholic acid (Tanaka
  and Makino, 1992; Sharma et al., 2011). However, genome-wide
  gene expression studies did not reveal any relevant and consistent
  changes in parkin-mutant fibroblasts after treatment with ursolic
  acid or DUA (data not shown). In particular, there was no effect
  on messenger RNA levels of mitochondrial master regulators such
  as PGC1alpha (now known as PPARGC1A) or mitochondrial
  uncoupling proteins. The biological function of glucocorticoids
  encompasses both genomic and non-genomic effects, including
  direct binding to the mitochondrial membrane, which can lead
  to partial uncoupling of oxidative phosphorylation (Haller et al.,
  2008).
  We appreciate that our work largely focused on assessing the
  effect of compounds in parkin-mutant Parkinson’s disease patient
  tissue. However, the beneficial effect of the lead compound, ursocholanic
  acid and the chemically related licensed drug ursodeoxycholic
  acid were also clearly apparent in LRRK2G2019S mutant
  fibroblasts. Ten per cent of all sporadic and 30% of familial
  Parkinson’s disease can be due to the LRRK2G2019S mutation in
  Ashkenazi Jewish patients with Parkinson’s disease (Ozelius et al.,
  2006). The prevalence may be even higher in other populations
  (Lesage et al., 2006). The mitochondrial phenotype is generally
  accepted to be correct for PARK2 but additional work is needed to
  determine whether rescue of mitochondrial function will result in
  at least partial rescue of neuronal dysfunction and cell loss in
  LRRK2G2019S-mutant model systems. If this was to be the case,
  then our lead compounds or structurally related drugs may already
  have a beneficial effect in a significant number of patients with
  Parkinson’s disease even if their effect was limited to parkin- and
  LRRK2G2019S mutant patients with Parkinson’s disease only.
  Mitochondrial dysfunction was first implicated in the pathogenesis
  of Parkinson’s disease when drug abusers developed parkinsonism
  after accidental exposure to the complex I inhibitor MPTP
  (Abou-Sleiman et al., 2006; Schapira, 2008). Subsequently, several
  groups reported independently decreased complex I activity in
  Parkinson’s disease (Mizuno et al., 1989; Parker et al., 1989;
  Schapira et al., 1989). It is now widely accepted that mitochondrial
  dysfunction and impaired morphology play a crucial role in
  the pathogenesis of early-onset Parkinson’s disease due to mutations
  in parkin (PARK2), PINK1 or DJ1 (PARK7) (Cookson and
  Bandmann, 2010). Mitochondrial dysfunction has also been
  observed in patient tissue (see above) or model systems of lateonset
  Parkinson’s disease due to mutations in LRRK2 or alpha
  synuclein (SNCA) (Loeb et al., 2010; Hindle et al., 2013). Akt, a
  protein kinase with multiple targets, is activated by successive
  phosphorylation at two sites. Failure of Akt signalling has been
  described as the ‘common core’ underlying neuronal degeneration
  and cell death in both familial and sporadic Parkinson’s disease
  (Greene et al., 2011). Akt phosphorylation is reduced in dopaminergic
  neurons of sporadic Parkinson’s disease (Malagelada et al.,
  2008; Timmons et al., 2009). Both increased expression of alpha
  synuclein (SNCA) and SNCA mutations lead to reduced Akt
  Figure 5 The rescue effect of ursocholanic acid and DUA is Akt
  mediated. (A) pAktSer473 protein levels as a ratio to total Akt
  protein levels as measured by ELISA. pAktSer473 levels are
  increased in parkin-mutant patient cells after treatment with
  both ursocholanic acid (grey bars) and DUA (black bars)
  (***P50.001, *P50.05). (B and C) Cellular ATP levels in
  control fibroblasts (white bars) and parkin-mutant fibroblasts
  (black bars). Pretreatment with the phosphatidylinositol
  3-kinases (PI 3-kinase) inhibitor LY29400 or triciribine, which
  selectively inhibit the cellular phosphorylation/activation of Akt,
  abolish the rescue effect of both ursocholanic acid (B) and DUA
  (C) (*P50.05). DMSO = dimethylsulphoxide.
  Ursocholanic acid in familial PD Brain 2013: Page 9 of 13 | 9
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  activation (Chung et al., 2011). Similarly, LRRK2 mutations (in
  particular G2019S) as well as Parkin, PINK1 and DJ1 deficiency
  result in decreased Akt phosphorylation (Yang et al., 2005; Fallon
  et al., 2006; Murata et al., 2011; Ohta et al., 2011). In contrast,
  the protective effect of beta-synuclein is mediated by increased
  Akt phosphorylation and increased parkin expression normalizes
  reduced Akt phosphorylation in MPTP-treated mice (Hashimoto
  et al., 2004; Yasuda et al., 2011). Further work is needed to determine
  whether the mitochondrial rescue effect and increased Akt
  phosphorylation at Ser473 after treatment with ursocholanic acid
  and DUA (as observed in our parkin-mutant fibroblast model) can
  also be observed in other forms and model systems of Parkinson’s
  disease.
  Ursodeoxycholic acid has been licensed for the treatment of
  patients with primary biliary cirrhosis since 1980. It is typically
  used at a dose of 10 mg/kg of body weight per day in patients
  with primary biliary cirrhosis but Parry et al. (2010) also reported
  ‘excellent’ safety and tolerability of ursodeoxycholic acid in
  patients with motor neuron disease at 15 mg, 30mg and 50 mg/
  kg per day. There was a significant correlation between serum and
  CSF concentrations of ursodeoxycholic acid. There is therefore
  good rationale to assume that ursodeoxycholic acid may also be
  well tolerated in Parkinson’s disease and cross the blood–brain
  barrier. Drug repositioning of FDA-licensed drugs such as ursodeoxycholic
  acid is a promising strategy to save time and costs
  but Parkinson’s disease-specific, reliable data on safety, tolerability
  and CSF penetration of ursodeoxycholic acid will nevertheless be
  of paramount importance before ursodeoxycholic acid can be
  taken into clinical trials to assess its putative disease-modifying
  effect in Parkinson’s disease.
  Dopaminergic neurons derived from inducible stem cells have
  already been used to assess compounds for their putative rescue
  effect on crucial pathogenic mechanisms for Parkinson’s disease
  and other conditions (Cooper et al., 2012). However, the inducible
  stem cells-based approach, although in many ways exciting and
  promising, is also costly and not without inherent problems. Our
  study demonstrates that a step-wise strategy, encompassing an
  initial screen in Parkinson’s disease patient fibroblasts but
  Figure 6 Rescue of cellular ATP levels by 24-h treatment of parkin-mutant fibroblasts with 100nM ursocholanic acid (UCA, A), DUA
  (B), ursolic acid (UA, C) and ursodeoxycholic acid (UDCA, D). Cellular ATP levels are significantly reduced in untreated parkin-mutant
  patient fibroblasts (*P50.05) but significantly increased after treatment with any of these four respective drugs (*P50.05, **P50.01).
  10 | Brain 2013: Page 10 of 13 H. Mortiboys et al.
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013
  subsequent confirmation of top hits in a neuronal model system
  may be a less costly and more robust strategy.
  Previous studies investigating the potential rescue effect of
  pharmacological compounds in model systems of early onset
  Parkinson’s disease have concentrated on a hypothesis-driven
  approach testing individual compounds rather than assessing a
  compound library in a hypothesis-free approach. Vitamin K(2)
  acts as a mitochondrial electron carrier that rescues mitochondrial
  dysfunction in pink1-deficient Drosophila (Vos et al., 2012).
  However, it is unclear whether vitamin K(2) also rescues mitochondrial
  dysfunction in Parkin deficiency. The disaccharide trehalose
  increases the removal of abnormal proteins through
  enhancement of autophagy. Trehalose treatment ameliorates tau
  pathology but fails to revert the loss of dopaminergic neurons in a
  mouse model of tauopathy with parkinsonism, overexpressing
  human mutated tau protein with deletion of parkin (Rodriguez-
  Navarro et al., 2010). Co-enzyme Q10 reduces the vulnerability of
  inducible stem cell-derived, PINK1 mutant neural cells to the
  lowest, but not to high concentrations of valinomycin and concamycin
  A, rapamycin did not reduce lactate dehydrogenase release
  after exposure to these toxins. In contrast, both rapamycin and
  the LRRK2 inhibitor GW5074 reduced the production of mitochondrial
  reactive oxygen species in PINK1 mutant neural cells
  exposed to valinomycin. However, none of these compounds
  were assessed for their rescue effect on baseline mitochondrial
  (dys)function in PINK1 mutant model systems before toxin exposure
  (Cooper et al., 2012). Future drug screens may be preceeded
  by in silico screens assessing compounds for their likely effect on
  enhancing the biological activity of proteins such as Parkin or
  PINK1, but also on other proteins such as thioredoxin with a
  reported rescue effect in Parkin-deficient Drosophila (Umeda-
  Kameyama et al., 2007; Trempe et al., 2013). Other therapeutic
  approaches include the overexpression of enzymes bypassing complex
  I activity such as the Saccaromyces cerevisiae enzyme Ndi1p
  (Vilain et al., 2012).
  Acknowledgements
  We would like to thank all research participants.
  Funding
  Financial support from Parkinson’s UK (G-0715 and G-0901) is
  gratefully acknowledged.
  Suppplementary material
  Supplementary material is available at Brain online.
  References
  Abou-Sleiman PM, Muqit MM, Wood NW. Expanding insights of mitochondrial
  dysfunction in Parkinson’s disease. Nat Rev Neurosci 2006;
  7: 207–19.
  Figure 7 Ursocholanic acid (UCA) and ursodeoxycholic acid
  (UDCA) rescue effect in cortical neurons with small interfering
  RNA mediated parkin knockdown. (A) Western blot showing
  Parkin band at 50 kDa and actin band at 40 kDa in scramble
  small interfering RNA and parkin small interfering RNA transfected
  cortical neurons. (B) Parkin protein levels are reduced by
  80% in parkin small interfering RNA knockdown cortical
  neurons as assessed by western blotting (***P50.001).
  (C) Cellular ATP levels in cortical neurons at 9 days in culture
  transfected with either scramble small interfering RNA (white
  bars), or parkin small interfering RNA (black bars). There is a
  reduction of 43% in cellular ATP levels in the parkin small
  interfering RNA transfected cells, (**P50.01), which is rescued
  by treatment with 10pM ursocholanic acid or ursodeoxycholic
  acid. DMSO = dimethylsulphoxide.
  Figure 8 Cellular ATP levels are reduced in fibroblasts from
  three LRRK2G2019S mutant patients (black bars) compared with
  controls (white bars) *P50.05. There is complete recovery of
  ATP to normal levels after treatment with 10nM ursocholanic
  acid or 10nM ursodeoxycholic acid for 24 h.
  DMSO = dimethylsulphoxide.
  Ursocholanic acid in familial PD Brain 2013: Page 11 of 13 | 11
  Downloaded from http://brain.oxfordjournals.org/ at Serials Department on September 9, 2013 

hyh883:回复·  邮箱:123456  IP:122.235.176.83  日期:2013-9-10 [回复18楼]

  回复·
  多谢薛老,看来大家没了薛老还真的不行!希薛老多保重身体,我想这是我们大家的心声 

崔亚:回复12楼 薛传恽  IP:222.187.192.150  日期:2013-9-10 [回复19楼]

  回复12楼 薛传恽
  回复12楼 薛传恽:回复1楼 薛传恽
  
  感动于薛老的钻研精神!
  
  感恩于薛老的无私奉献!
  
  感谢薛老的大善之举! 

summer:回复1楼 薛传恽  邮箱:blueapple406@163.com  IP:60.30.4.34  日期:2013-9-10 [回复20楼]

  回复1楼 薛传恽
  回复1楼 薛传恽:令人兴奋的新消息
  
  还需要和医生商量吗?可以直接吃吗?我都不知道和哪个医生商量,因为从我母亲确诊后,就去过一次医院。我都是按您三步走的宗旨吃药的,目前还是只吃雷。虽然症状无缓解,但母亲一直能坚持!
  
  离不开您,看来是很多病友的共识了~~薛老师,您多保重哦! 

输1-2个字  第 1 2 3 4 5 6 7 8 9 10 31 页(共306条)