SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "AMNE:(MEDICIN OCH HÄLSOVETENSKAP Medicinska och farmaceutiska grundvetenskaper Farmaceutiska vetenskaper) ;pers:(Uhlen Mathias)"

Sökning: AMNE:(MEDICIN OCH HÄLSOVETENSKAP Medicinska och farmaceutiska grundvetenskaper Farmaceutiska vetenskaper) > Uhlen Mathias

  • Resultat 1-10 av 17
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Turanli, Beste, et al. (författare)
  • Drug Repositioning for Effective Prostate Cancer Treatment
  • 2018
  • Ingår i: Frontiers in Physiology. - : Frontiers Media SA. - 1664-042X. ; 9
  • Tidskriftsartikel (refereegranskat)abstract
    • Drug repositioning has gained attention from both academia and pharmaceutical companies as an auxiliary process to conventional drug discovery. Chemotherapeutic agents have notorious adverse effects that drastically reduce the life quality of cancer patients so drug repositioning is a promising strategy to identify non-cancer drugs which have anti-cancer activity as well as tolerable adverse effects for human health. There are various strategies for discovery and validation of repurposed drugs. In this review, 25 repurposed drug candidates are presented as result of different strategies, 15 of which are already under clinical investigation for treatment of prostate cancer (PCa). To date, zoledronic acid is the only repurposed, clinically used, and approved non-cancer drug for PCa. Anti-cancer activities of existing drugs presented in this review cover diverse and also known mechanisms such as inhibition of mTOR and VEGFR2 signaling, inhibition of PI3K/Akt signaling, COX and selective COX-2 inhibition, NF-kappa B inhibition, Wnt/beta - Catenin pathway inhibition, DNMT1 inhibition, and GSK-3 beta inhibition. In addition to monotherapy option, combination therapy with current anti-cancer drugs may also increase drug efficacy and reduce adverse effects. Thus, drug repositioning may become a key approach for drug discovery in terms of time- and cost-efficiency comparing to conventional drug discovery and development process.
  •  
2.
  • Mohammadi, Elyas, et al. (författare)
  • Applications of Genome-Wide Screening and Systems Biology Approaches in Drug Repositioning
  • 2020
  • Ingår i: Cancers. - : MDPI AG. - 2072-6694. ; 12:9, s. 1-24
  • Forskningsöversikt (refereegranskat)abstract
    • Simple Summary Drug repurposing is an accelerated route for drug development and a promising approach for finding medications for orphan and common diseases. Here, we compiled databases that comprise both computationally- or experimentally-derived data, and categorized them based on quiddity and origin of data, further focusing on those that present high throughput omic data or drug screens. These databases were then contextualized with genome-wide screening methods such as CRISPR/Cas9 and RNA interference, as well as state of art systems biology approaches that enable systematic characterizations of multi-omic data to find new indications for approved drugs or those that reached the latest phases of clinical trials. Modern drug discovery through de novo drug discovery entails high financial costs, low success rates, and lengthy trial periods. Drug repositioning presents a suitable approach for overcoming these issues by re-evaluating biological targets and modes of action of approved drugs. Coupling high-throughput technologies with genome-wide essentiality screens, network analysis, genome-scale metabolic modeling, and machine learning techniques enables the proposal of new drug-target signatures and uncovers unanticipated modes of action for available drugs. Here, we discuss the current issues associated with drug repositioning in light of curated high-throughput multi-omic databases, genome-wide screening technologies, and their application in systems biology/medicine approaches.
  •  
3.
  • Altay, Özlem, et al. (författare)
  • Current Status of COVID-19 Therapies and Drug Repositioning Applications
  • 2020
  • Ingår i: Iscience. - : Elsevier BV. - 2589-0042. ; 23:7
  • Tidskriftsartikel (refereegranskat)abstract
    • The rapid and global spread of a new human coronavirus (SARS-CoV-2) has produced an immediate urgency to discover promising targets for the treatment of COVID-19. Drug repositioning is an attractive approach that can facilitate the drug discovery process by repurposing existing pharmaceuticals to treat illnesses other than their primary indications. Here, we review current information concerning the global health issue of COVID-19 including promising approved drugs and ongoing clinical trials for prospective treatment options. In addition, we describe computational approaches to be used in drug repurposing and highlight examples of in silico studies of drug development efforts against SARS-CoV-2.
  •  
4.
  • Zhang, Cheng, et al. (författare)
  • Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositioning
  • 2022
  • Ingår i: eBioMedicine. - : Elsevier BV. - 2352-3964. ; 83
  • Tidskriftsartikel (refereegranskat)abstract
    • Background Non-alcoholic fatty liver disease (NAFLD) encompasses a wide spectrum of liver pathologies. However, no medical treatment has been approved for the treatment of NAFLD. In our previous study, we found that PKLR could be a potential target for treatment of NALFD. Here, we investigated the effect of PKLR in in vivo model and performed drug repositioning to identify a drug candidate for treatment of NAFLD. Methods Tissue samples from liver, muscle, white adipose and heart were obtained from control and PKLR knock-out mice fed with chow and high sucrose diets. Lipidomics as well as transcriptomics analyses were conducted using these tissue samples. In addition, a computational drug repositioning analysis was performed and drug candidates were identified. The drug candidates were both tested in in vitro and in vivo models to evaluate their toxicity and efficacy. Findings The Pklr KO reversed the increased hepatic triglyceride level in mice fed with high sucrose diet and partly recovered the transcriptomic changes in the liver as well as in other three tissues. Both liver and white adipose tissues exhibited dysregulated circadian transcriptomic profiles, and these dysregulations were reversed by hepatic knockout of Pklr. In addition, 10 small molecule drug candidates were identified as potential inhibitor of PKLR using our drug repositioning pipeline, and two of them significantly inhibited both the PKLR expression and triglyceride level in in vitro model. Finally, the two selected small molecule drugs were evaluated in in vivo rat models and we found that these drugs attenuate the hepatic steatosis without side effect on other tissues. Interpretation In conclusion, our study provided biological insights about the critical role of PKLR in NAFLD progression and proposed a treatment strategy for NAFLD patients, which has been validated in preclinical studies. Copyright (C) 2022 The Authors. Published by Elsevier B.V.
  •  
5.
  • Zhang, C., et al. (författare)
  • The acute effect of metabolic cofactor supplementation: a potential therapeutic strategy against non-alcoholic fatty liver disease
  • 2020
  • Ingår i: Molecular Systems Biology. - : EMBO. - 1744-4292. ; 16:4
  • Tidskriftsartikel (refereegranskat)abstract
    • The prevalence of non-alcoholic fatty liver disease (NAFLD) continues to increase dramatically, and there is no approved medication for its treatment. Recently, we predicted the underlying molecular mechanisms involved in the progression of NAFLD using network analysis and identified metabolic cofactors that might be beneficial as supplements to decrease human liver fat. Here, we first assessed the tolerability of the combined metabolic cofactors including l-serine, N-acetyl-l-cysteine (NAC), nicotinamide riboside (NR), and l-carnitine by performing a 7-day rat toxicology study. Second, we performed a human calibration study by supplementing combined metabolic cofactors and a control study to study the kinetics of these metabolites in the plasma of healthy subjects with and without supplementation. We measured clinical parameters and observed no immediate side effects. Next, we generated plasma metabolomics and inflammatory protein markers data to reveal the acute changes associated with the supplementation of the metabolic cofactors. We also integrated metabolomics data using personalized genome-scale metabolic modeling and observed that such supplementation significantly affects the global human lipid, amino acid, and antioxidant metabolism. Finally, we predicted blood concentrations of these compounds during daily long-term supplementation by generating an ordinary differential equation model and liver concentrations of serine by generating a pharmacokinetic model and finally adjusted the doses of individual metabolic cofactors for future human clinical trials.
  •  
6.
  • Zeybel, M., et al. (författare)
  • Multiomics Analysis Reveals the Impact of Microbiota on Host Metabolism in Hepatic Steatosis
  • 2022
  • Ingår i: Advanced Science. - : Wiley. - 2198-3844. ; 9:11, s. 2104373-
  • Tidskriftsartikel (refereegranskat)abstract
    • Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex disease involving alterations in multiple biological processes regulated by the interactions between obesity, genetic background, and environmental factors including the microbiome. To decipher hepatic steatosis (HS) pathogenesis by excluding critical confounding factors including genetic variants and diabetes, 56 heterogenous MAFLD patients are characterized by generating multiomics data including oral and gut metagenomics as well as plasma metabolomics and inflammatory proteomics data. The dysbiosis in the oral and gut microbiome is explored and the host–microbiome interactions based on global metabolic and inflammatory processes are revealed. These multiomics data are integrated using the biological network and HS's key features are identified using multiomics data. HS is finally predicted using these key features and findings are validated in a follow-up cohort, where 22 subjects with varying degree of HS are characterized.
  •  
7.
  • Bosley, J. R., et al. (författare)
  • Informing Pharmacokinetic Models With Physiological Data: Oral Population Modeling of L-Serine in Humans
  • 2021
  • Ingår i: Frontiers in Pharmacology. - : Frontiers Media SA. - 1663-9812. ; 12
  • Tidskriftsartikel (refereegranskat)abstract
    • To determine how to set optimal oral L-serine (serine) dose levels for a clinical trial, existing literature was surveyed. Data sufficient to set the dose was inadequate, and so an (n = 10) phase I-A calibration trial was performed, administering serine with and without other oral agents. We analyzed the trial and the literature data using pharmacokinetic (PK) modeling and statistical analysis. The therapeutic goal is to modulate specific serine-related metabolic pathways in the liver using the lowest possible dose which gives the desired effect since the upper bound was expected to be limited by toxicity. A standard PK approach, in which a common model structure was selected using a fit to data, yielded a model with a single central compartment corresponding to plasma, clearance from that compartment, and an endogenous source of serine. To improve conditioning, a parametric structure was changed to estimate ratios (bioavailability over volume, for example). Model fit quality was improved and the uncertainty in estimated parameters was reduced. Because of the particular interest in the fate of serine, the model was used to estimate whether serine is consumed in the gut, absorbed by the liver, or entered the blood in either a free state, or in a protein- or tissue-bound state that is not measured by our assay. The PK model structure was set up to represent relevant physiology, and this quantitative systems biology approach allowed a broader set of physiological data to be used to narrow parameter and prediction confidence intervals, and to better understand the biological meaning of the data. The model results allowed us to determine the optimal human dose for future trials, including a trial design component including IV and tracer studies. A key contribution is that we were able to use human physiological data from the literature to inform the PK model and to set reasonable bounds on parameters, and to improve model conditioning. Leveraging literature data produced a more predictive, useful model.
  •  
8.
  • Li, Xiangyu, et al. (författare)
  • Prediction of drug candidates for clear cell renal cell carcinoma using a systems biology-based drug repositioning approach
  • 2022
  • Ingår i: EBioMedicine. - : Elsevier BV. - 2352-3964. ; 78, s. 103963-
  • Tidskriftsartikel (refereegranskat)abstract
    • SummaryBackground: The response rates of the clinical chemotherapies are still low in clear cell renal cell carcinoma (ccRCC). Computational drug repositioning is a promising strategy to discover new uses for existing drugs to treat patients who cannot get benefits from clinical drugs.Methods: We proposed a systematic approach which included the target prediction based on the co-expression network analysis of transcriptomics profiles of ccRCC patients and drug repositioning for cancer treatment based on the analysis of shRNA-and drug-perturbed signature profiles of human kidney cell line.Findings: First, based on the gene co-expression network analysis, we identified two types of gene modules in ccRCC, which significantly enriched with unfavorable and favorable signatures indicating poor and good survival outcomes of patients, respectively. Then, we selected four genes, BUB1B, RRM2, ASF1B and CCNB2, as the potential drug targets based on the topology analysis of modules. Further, we repurposed three most effective drugs for each target by applying the proposed drug repositioning approach. Finally, we evaluated the effects of repurposed drugs using an in vitro model and observed that these drugs inhibited the protein levels of their corresponding target genes and cell viability.Interpretation: These findings proved the usefulness and efficiency of our approach to improve the drug repositioning researches for cancer treatment and precision medicine.Funding: This study was funded by Knut and Alice Wallenberg Foundation and Bash Biotech Inc., San Diego, CA, USA. 
  •  
9.
  • Turanli, Beste Calimlioglu, et al. (författare)
  • Systems biology based drug repositioning for development of cancer therapy
  • 2021
  • Ingår i: Seminars in Cancer Biology. - : Elsevier BV. - 1096-3650 .- 1044-579X. ; 68, s. 47-58
  • Forskningsöversikt (refereegranskat)abstract
    • Drug repositioning is a powerful method that can assists the conventional drug discovery process by using existing drugs for treatment of a disease rather than its original indication. The first examples of repurposed drugs were discovered serendipitously, however data accumulated by high-throughput screenings and advancements in computational biology methods have paved the way for rational drug repositioning methods. As chemotherapeutic agents have notorious side effects that significantly reduce quality of life, drug repositioning promises repurposed noncancer drugs with little or tolerable adverse effects for cancer patients. Here, we review current drug-related data types and databases including some examples of web-based drug repositioning tools. Next, we describe systems biology approaches to be used in drug repositioning for effective cancer therapy. Finally, we highlight examples of mostly repurposed drugs for cancer treatment and provide an overview of future expectations in the field for development of effective treatment strategies.
  •  
10.
  • Mardinoglu, Adil, 1982, et al. (författare)
  • Personal model-assisted identification of NAD(+) and glutathione metabolism as intervention target in NAFLD
  • 2017
  • Ingår i: Molecular Systems Biology. - : EMBO. - 1744-4292. ; 13:3
  • Tidskriftsartikel (refereegranskat)abstract
    • To elucidate the molecular mechanisms underlying non-alcoholic fatty liver disease (NAFLD), we recruited 86 subjects with varying degrees of hepatic steatosis (HS). We obtained experimental data on lipoprotein fluxes and used these individual measurements as personalized constraints of a hepatocyte genome-scale metabolic model to investigate metabolic differences in liver, taking into account its interactions with other tissues. Our systems level analysis predicted an altered demand for NAD(+) and glutathione (GSH) in subjects with high HS. Our analysis and metabolomic measurements showed that plasma levels of glycine, serine, and associated metabolites are negatively correlated with HS, suggesting that these GSH metabolism precursors might be limiting. Quantification of the hepatic expression levels of the associated enzymes further pointed to altered de novo GSH synthesis. To assess the effect of GSH and NAD(+) repletion on the development of NAFLD, we added precursors for GSH and NAD(+) biosynthesis to the Western diet and demonstrated that supplementation prevents HS in mice. In a proof-of-concept human study, we found improved liver function and decreased HS after supplementation with serine (a precursor to glycine) and hereby propose a strategy for NAFLD treatment.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 17
Typ av publikation
tidskriftsartikel (15)
forskningsöversikt (2)
Typ av innehåll
refereegranskat (17)
Författare/redaktör
Uhlén, Mathias (17)
Nielsen, Jens B, 196 ... (12)
Borén, Jan, 1963 (11)
Mardinoglu, Adil, 19 ... (10)
Turkez, Hasan (6)
Zhang, C. (6)
visa fler...
Kim, Woonghee (6)
Mardinoglu, Adil (5)
Li, Xiangyu (5)
Turkez, H. (5)
Zhang, Cheng (4)
Benfeitas, Rui (4)
Altay, Özlem (4)
Arif, Muhammad (3)
Björnson, Elias, 198 ... (3)
Yang, Hong (3)
Marschall, Hanns-Ulr ... (2)
Adiels, Martin, 1976 (2)
Bidkhori, Gholamreza (2)
Ståhlman, Marcus, 19 ... (2)
Shoaie, Saeed (2)
Mohammadi, Elyas (2)
Lam, S. (2)
Klevstig, Martina (2)
Al-Khalili Szigyarto ... (1)
Pontén, Fredrik (1)
Abdellah, Tebani (1)
Achour, Adnane (1)
Rodriguez, Henry (1)
Ping, Peipei (1)
Taskinen, Marja-Riit ... (1)
Taskinen, M. R. (1)
Soderlund, S (1)
Hakkarainen, A. (1)
Lundbom, N. (1)
Karlsson, Johan (1)
Grötli, Morten (1)
Mallick, Parag (1)
Ahlin, Gustav, 1977- (1)
Hilgendorf, Constanz ... (1)
Artursson, Per (1)
Dunn, Michael (1)
Smith, Ulf, 1943 (1)
Hallstrom, B. M. (1)
Apweiler, Rolf (1)
Doran, S (1)
Ashraf, Sajda (1)
Bergh, Per-Olof (1)
Hermjakob, Henning (1)
Iqbal, S (1)
visa färre...
Lärosäte
Kungliga Tekniska Högskolan (17)
Chalmers tekniska högskola (14)
Göteborgs universitet (11)
Karolinska Institutet (4)
Uppsala universitet (2)
Stockholms universitet (2)
Språk
Engelska (17)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (17)
Naturvetenskap (10)
Teknik (1)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy