Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Publications (10 of 27) Show all publications
Mickols, E., Mohammedamin, R., Primpas, L., Oredsson, S. & Karlgren, M. (2025). Animal product–free formation and cultivation of three-dimensional primary hepatocyte spheroids. Drug Metabolism And Disposition, 53(10), Article ID 100147.
Open this publication in new window or tab >>Animal product–free formation and cultivation of three-dimensional primary hepatocyte spheroids
Show others...
2025 (English)In: Drug Metabolism And Disposition, ISSN 0090-9556, E-ISSN 1521-009X, Vol. 53, no 10, article id 100147Article in journal (Refereed) Published
Abstract [en]

Three-dimensional (3D) cultures of primary human hepatocytes (3D PHH) are successfully used to reduce and replace the use of animal experiments in biomedical research. Yet, the initial formation of 3D PHH is highly dependent on the supplementation with FBS. However, the molecular composition of FBS and its effects on cultured cells are poorly understood. Moreover, FBS is prone to batch-to-batch variation, immunogenic risk, and lack of adherence to the replacement, refinement, and reduction of animal experiments. Here, we demonstrate that FBS can be fully replaced by animal-free substitutes, thus facilitating fully chemically defined and animal serum–free 3D PHH cultures. Specifically, we combined a previously developed animal-free substitute cocktail with a normoglycemic (5.5 mM glucose and 0.58 ng/mL insulin) chemically defined culture medium. Morphological and viability evaluations, along with global proteomics data, demonstrated that serum-free cultured 3D PHH have comparable viability and functional performance of cytochrome P450s, rendering this medium useful for long-term studies and in vitro absorption, distribution, metabolism, excretion, and toxicity applications. This study marks a significant advancement in the development of animal serum–free culture conditions for primary human cell cultures, paving the way for more reliable and ethical in vitro studies.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
3D PHH, Primary human hepatocytes, Fetal bovine serum, Serum-free, In vitro model
National Category
Pharmaceutical and Medical Biotechnology Pharmacology and Toxicology
Identifiers
urn:nbn:se:uu:diva-548556 (URN)10.1016/j.dmd.2025.100147 (DOI)001582160100002 ()40945386 (PubMedID)2-s2.0-105015560959 (Scopus ID)
Note

Authors in the list of papers of Evgeniya Mickols thesis: Mickols, E., Primpas, L., Oredsson, S., Karlgren, M.

Available from: 2025-01-27 Created: 2025-01-27 Last updated: 2025-10-10Bibliographically approved
Mickols, E., Meyer, A., Handin, N., Stüwe, M., Eriksson, J., Rudfeldt, J., . . . Artursson, P. (2024). OCT1 (SLC22A1) transporter kinetics and regulation in primary human hepatocyte 3D spheroids. Scientific Reports, 14(1), Article ID 17334.
Open this publication in new window or tab >>OCT1 (SLC22A1) transporter kinetics and regulation in primary human hepatocyte 3D spheroids
Show others...
2024 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 14, no 1, article id 17334Article in journal (Refereed) Published
Abstract [en]

3D spheroids of primary human hepatocytes (3D PHH) retain a differentiated phenotype with largely conserved metabolic function and proteomic fingerprint over weeks in culture. As a result, 3D PHH are gaining importance as a model for mechanistic liver homeostasis studies and in vitro to in vivo extrapolation (IVIVE) in drug discovery. However, the kinetics and regulation of drug transporters have not yet been assessed in 3D PHH. Here, we used organic cation transporter 1 (OCT1/SLC22A1) as a model to study both transport kinetics and the long-term regulation of transporter activity via relevant signalling pathways. The kinetics of the OCT1 transporter was studied using the fluorescent model substrate 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+) and known OCT1 inhibitors in individual 3D PHH. For long-term studies, 3D PHH were treated with xenobiotics for seven days, after which protein expression and OCT1 function were assessed. Global proteomic analysis was used to track hepatic phenotypes as well as prototypical changes in other regulated proteins, such as P-glycoprotein and Cytochrome P450 3A4. ASP+ kinetics indicated a fully functional OCT1 transporter with a Km value of 14 ± 4.0µM as the mean from three donors. Co-incubation with known OCT1 inhibitors decreased the uptake of ASP+ in the 3D PHH spheroids by 35–52%. The long-term exposure studies showed that OCT1 is relatively stable upon activation of nuclear receptor signalling or exposure to compounds that could induce inflammation, steatosis or liver injury. Our results demonstrate that 3D PHH spheroids express physiologically relevant levels of fully active OCT1 and that its transporter kinetics can be accurately studied in the 3D PHH configuration. We also confirm that OCT1 remains stable and functional during the activation of key metabolic pathways that alter the expression and function of other drug transporters and drug-metabolizing enzymes. These results will expand the range of studies that can be performed using 3D PHH.

Place, publisher, year, edition, pages
Springer Nature, 2024
Keywords
Hepatocyte, Liver, Uptake, ASP+, OCT1, Drug transport, Drug-Drug interaction, Spheroid, 3D culture, Proteomics
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-535505 (URN)10.1038/s41598-024-67192-6 (DOI)001281273100063 ()39068198 (PubMedID)2-s2.0-85199759857 (Scopus ID)
Funder
Uppsala UniversitySwedish Research Council, 2020-01586Swedish Research Council, 2017-01951
Note

These authors contributed equally: Evgeniya Mickols, Alina Meyer and Niklas Handin.

Available from: 2024-08-01 Created: 2024-08-01 Last updated: 2025-02-13Bibliographically approved
Rahman Ansari, S., Suárez-López, Y. d., Thersleff, T., Häggström, L., Ericsson, T., Katsaros, I., . . . Teleki, A. (2024). Pharmaceutical Quality by Design Approach to Develop High-Performance Nanoparticles for Magnetic Hyperthermia. ACS Nano, 18(23), 15284-15302
Open this publication in new window or tab >>Pharmaceutical Quality by Design Approach to Develop High-Performance Nanoparticles for Magnetic Hyperthermia
Show others...
2024 (English)In: ACS Nano, ISSN 1936-0851, E-ISSN 1936-086X, Vol. 18, no 23, p. 15284-15302Article in journal (Refereed) Published
Abstract [en]

Magnetic hyperthermia holds significant therapeutic potential, yet its clinical adoption faces challenges. One obstacle is the large-scale synthesis of high-quality superparamagnetic iron oxide nanoparticles (SPIONs) required for inducing hyperthermia. Robust and scalable manufacturing would ensure control over the key quality attributes of SPIONs, and facilitate clinical translation and regulatory approval. Therefore, we implemented a risk-based pharmaceutical quality by design (QbD) approach for SPION production using flame spray pyrolysis (FSP), a scalable technique with excellent batch-to-batch consistency. A design of experiments method enabled precise size control during manufacturing. Subsequent modeling linked the SPION size (6–30 nm) and composition to intrinsic loss power (ILP), a measure of hyperthermia performance. FSP successfully fine-tuned the SPION composition with dopants (Zn, Mn, Mg), at various concentrations. Hyperthermia performance showed a strong nonlinear relationship with SPION size and composition. Moreover, the ILP demonstrated a stronger correlation to coercivity and remanence than to the saturation magnetization of SPIONs. The optimal operating space identified the midsized (15–18 nm) Mn0.25Fe2.75O4 as the most promising nanoparticle for hyperthermia. The production of these nanoparticles on a pilot scale showed the feasibility of large-scale manufacturing, and cytotoxicity investigations in multiple cell lines confirmed their biocompatibility. In vitro hyperthermia studies with Caco-2 cells revealed that Mn0.25Fe2.75O4 nanoparticles induced 80% greater cell death than undoped SPIONs. The systematic QbD approach developed here incorporates process robustness, scalability, and predictability, thus, supporting the clinical translation of high-performance SPIONs for magnetic hyperthermia.

Place, publisher, year, edition, pages
American Chemical Society (ACS), 2024
Keywords
quality by design, superparamagnetic nanoparticles, magnetic hyperthermia, design of experiments, flame spray pyrolysis, doped ferrites
National Category
Pharmaceutical Sciences Other Materials Engineering
Research subject
Pharmaceutical Science
Identifiers
urn:nbn:se:uu:diva-527076 (URN)10.1021/acsnano.4c04685 (DOI)001236198600001 ()38814737 (PubMedID)
Funder
Science for Life Laboratory, SciLifeLabEU, Horizon 2020, 101002582
Note

Title in the list of papers of Shaquib Rahman Ansari's thesis: A pharmaceutical quality by design approach to develop high performance nanoparticles for magnetic hyperthermia

Available from: 2024-04-23 Created: 2024-04-23 Last updated: 2024-10-24Bibliographically approved
Wegler, C., Gazit, M., Issa, K., Subramaniam, S., Artursson, P. & Karlgren, M. (2021). Expanding the Efflux In Vitro Assay Toolbox: A CRISPR-Cas9 Edited MDCK Cell Line with Human BCRP and Completely Lacking Canine MDR1. Journal of Pharmaceutical Sciences, 110(1), 388-396
Open this publication in new window or tab >>Expanding the Efflux In Vitro Assay Toolbox: A CRISPR-Cas9 Edited MDCK Cell Line with Human BCRP and Completely Lacking Canine MDR1
Show others...
2021 (English)In: Journal of Pharmaceutical Sciences, ISSN 0022-3549, E-ISSN 1520-6017, Vol. 110, no 1, p. 388-396Article in journal (Refereed) Published
Abstract [en]

The Breast Cancer Resistance Protein (BCRP) is a key transporter in drug efflux and drug-drug interactions. However, endogenous expression of Multidrug Resistance Protein 1 (MDR1) confounds the interpretation of BCRP-mediated transport in in vitro models. Here we used a CRISPR-Cas9 edited Madin-Darby canine kidney (MDCK) II cell line (MDCKcMDR1-KO) for stable expression of human BCRP (hBCRP) with no endogenous canine MDR1 (cMDR1) expression (MDCK-hBCRPcMDR1-KO). Targeted quantitative proteomics verified expression of hBCRP, and global analysis of the entire proteome corroborated no or very low background expression of other drug transport proteins or metabolizing enzymes. This new cell line, had similar proteome like MDCKcMDR1-KO and a previously established, corresponding cell line overexpressing human MDR1 (hMDR1), MDCK-hMDR1cMDR1-KO. Functional studies with MDCK-hBCRPcMDR1-KO confirmed high hBCRP activity. The MDCK-hBCRPcMDR1-KO cell line together with the MDCK-hMDR1cMDR1-KO easily and accurately identified shared or specific substrates of the hBCRP and the hMDR1 transporters. These cell lines offer new, improved in vitro tools for the assessment of drug efflux and drug-drug interactions in drug development.

Place, publisher, year, edition, pages
Elsevier, 2021
Keywords
ATP-binding cassette (ABC) transporter(s), Breast cancer resistance protein (BCRP), Drug transport, Efflux pumps, MDCK cells, Membrane transporter, P-glycoprotein (P-gp), Permeability, Proteomics
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-423990 (URN)10.1016/j.xphs.2020.09.039 (DOI)000600571800037 ()33007277 (PubMedID)
Funder
Swedish Fund for Research Without Animal Experiments, F2017-0004Swedish Fund for Research Without Animal Experiments, F2018-0004Swedish Fund for Research Without Animal Experiments, F2019-0001Magnus Bergvall Foundation, 2017-02305Magnus Bergvall Foundation, 2018-02721Åke Wiberg Foundation, M16-0074Swedish Research Council, 5715Swedish Research Council, 01951
Available from: 2020-10-30 Created: 2020-10-30 Last updated: 2024-01-15Bibliographically approved
Vallianatou, T., Shariatgorji, R., Nilsson, A., Karlgren, M., Hulme, H., Fridjonsdottir, E., . . . Andrén, P. E. (2021). Integration of Mass Spectrometry Imaging and Machine Learning Visualizes Region-Specific Age-Induced and Drug-Target Metabolic Perturbations in the Brain. ACS Chemical Neuroscience, 12(10), 1811-1823
Open this publication in new window or tab >>Integration of Mass Spectrometry Imaging and Machine Learning Visualizes Region-Specific Age-Induced and Drug-Target Metabolic Perturbations in the Brain
Show others...
2021 (English)In: ACS Chemical Neuroscience, E-ISSN 1948-7193, Vol. 12, no 10, p. 1811-1823Article in journal (Refereed) Published
Abstract [en]

Detailed metabolic imaging of specific brain regions in early aging may expose pathophysiological mechanisms and indicate effective neuropharmacological targets in the onset of cognitive decline. Comprehensive imaging of brain aging and drug-target effects is restricted using conventional methodology. We simultaneously visualized multiple metabolic alterations induced by normal aging in specific regions of mouse brains by integrating Fourier-transform ion cyclotron resonance mass spectrometry imaging and combined supervised and unsupervised machine learning models. We examined the interplay between aging and the response to tacrine-induced acetylcholinesterase inhibition, a well-characterized therapeutic treatment against dementia. The dipeptide carnosine (β-alanyl-l-histidine) and the vitamin α-tocopherol were significantly elevated by aging in different brain regions. l-Carnitine and acetylcholine metabolism were found to be major pathways affected by aging and tacrine administration in a brain region-specific manner, indicating altered mitochondrial function and neurotransmission. The highly interconnected hippocampus and retrosplenial cortex displayed different age-induced alterations in lipids and acylcarnitines, reflecting diverse region-specific metabolic effects. The subregional differences observed in the hippocampal formation of several lipid metabolites demonstrate the unique potential of the technique compared to standard mass spectrometry approaches. An age-induced increase of endogenous antioxidants, such as α-tocopherol, in the hippocampus was detected, suggesting an augmentation of neuroprotective mechanisms in early aging. Our comprehensive imaging approach visualized heterogeneous age-induced metabolic perturbations in mitochondrial function, neurotransmission, and lipid signaling, not always attenuated by acetylcholinesterase inhibition.

Place, publisher, year, edition, pages
American Chemical Society (ACS), 2021
Keywords
Acetylcholine, aging, lipids, mass spectrometry imaging, brain metabolomics, tacrine
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-446894 (URN)10.1021/acschemneuro.1c00103 (DOI)000655248600007 ()33939923 (PubMedID)
Funder
EU, FP7, Seventh Framework Programme, 607517Swedish Foundation for Strategic Research , RIF14-0078Swedish Research Council, 2018-03320Swedish Research Council, 2018-05501The Swedish Brain Foundation, FO20180292Science for Life Laboratory - a national resource center for high-throughput molecular bioscience
Available from: 2021-06-28 Created: 2021-06-28 Last updated: 2024-01-15Bibliographically approved
Antonescu, I. E., Karlgren, M., Pedersen, M. L., Simoff, I., Bergström, C., Neuhoff, S., . . . Nielsen, C. U. (2020). Acamprosate Is a Substrate of the Human Organic Anion Transporter (OAT) 1 without OAT3 Inhibitory Properties: Implications for Renal Acamprosate Secretion and Drug-Drug Interactions. Pharmaceutics, 12(4), Article ID 390.
Open this publication in new window or tab >>Acamprosate Is a Substrate of the Human Organic Anion Transporter (OAT) 1 without OAT3 Inhibitory Properties: Implications for Renal Acamprosate Secretion and Drug-Drug Interactions
Show others...
2020 (English)In: Pharmaceutics, E-ISSN 1999-4923, Vol. 12, no 4, article id 390Article in journal (Refereed) Published
Abstract [en]

Acamprosate is an anionic drug substance widely used in treating symptoms of alcohol withdrawal. It was recently shown that oral acamprosate absorption is likely due to paracellular transport. In contrast, little is known about the eliminating mechanism clearing acamprosate from the blood in the kidneys, despite the fact that studies have shown renal secretion of acamprosate. The hypothesis of the present study was therefore that renal organic anion transporters (OATs) facilitate the renal excretion of acamprosate in humans. The aim of the present study was to establish and apply OAT1 (gene product of SLC22A6) and OAT3 (gene product of SLC22A8) expressing cell lines to investigate whether acamprosate is a substrate or inhibitor of OAT1 and/or OAT3. The studies were performed in HEK293-Flp-In cells stably transfected with SLC22A6 or SLC22A8. Protein and functional data showed that the established cell lines are useful for studying OAT1- and OAT3-mediated transport in bi-laboratory studies. Acamprosate inhibited OAT1-mediated p-aminohippuric acid (PAH) uptake but did not inhibit substrate uptake via OAT3 expressing cells, neither when applied concomitantly nor after a 3 h preincubation with acamprosate. The uptake of PAH via OAT1 was inhibited in a competitive manner by acamprosate and cellular uptake studies showed that acamprosate is a substrate for OAT1 with a K-m-value of approximately 700 mu M. Probenecid inhibited OAT1-mediated acamprosate uptake with a K-i-value of approximately 13 mu M, which may translate into an estimated clinically significant DDI index. In conclusion, acamprosate was identified as a substrate of OAT1 but not OAT3.

Place, publisher, year, edition, pages
MDPI, 2020
Keywords
acamprosate, probenecid, organic anion transporter, OAT1, OAT3, HEK293 cells, renal carrier, secretion, drug-drug interaction
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-413904 (URN)10.3390/pharmaceutics12040390 (DOI)000535575000095 ()32344570 (PubMedID)
Funder
Swedish Research Council, 01951NordForsk, 85352
Available from: 2020-06-24 Created: 2020-06-24 Last updated: 2024-07-04Bibliographically approved
Antonescu, I. E., Rasmussen, K. F., Neuhoff, S., Fretté, X., Karlgren, M., Bergström, C., . . . Steffansen, B. (2019). The Permeation of Acamprosate Is Predominantly Caused by Paracellular Diffusion across Caco-2 Cell Monolayers: A Paracellular Modeling Approach. Molecular Pharmaceutics, 16(11), 4636-4650
Open this publication in new window or tab >>The Permeation of Acamprosate Is Predominantly Caused by Paracellular Diffusion across Caco-2 Cell Monolayers: A Paracellular Modeling Approach
Show others...
2019 (English)In: Molecular Pharmaceutics, ISSN 1543-8384, E-ISSN 1543-8392, Vol. 16, no 11, p. 4636-4650Article in journal (Refereed) Published
Abstract [en]

In drug development, estimating fraction absorbed (Fa) in man for permeability-limited compounds is important but challenging. To model Fa of such compounds from apparent permeabilities (Papp) across filter-grown Caco-2 cell monolayers, it is central to elucidate the intestinal permeation mechanism(s) of the compound. The present study aims to refine a computational permeability model to investigate the relative contribution of paracellular and transcellular routes to the Papp across Caco-2 monolayers of the permeability-limited compound acamprosate having a bioavailability of ∼11%. The Papp values of acamprosate and of several paracellular marker molecules were measured. These Papp values were used to refine system-specific parameters of the Caco-2 monolayers, that is, paracellular pore radius, pore capacity, and potential drop. The refined parameters were subsequently used as an input in modeling the permeability (Pmodeled) of the tested compounds using mathematical models collected from two published permeability models. The experimental data show that acamprosate Papp across Caco-2 monolayers is low and similar in both transport directions. The obtained acamprosate Papp, 1.56 ± 0.28 × 10-7 cm·s-1, is similar to the Papp of molecular markers for paracellular permeability, namely, mannitol (2.72 ± 0.24 × 10-7 cm·s-1), lucifer yellow (1.80 ± 0.35 × 10-7 cm·s-1), and fluorescein (2.10 ± 0.28 × 10-7 cm·s-1), and lower than that of atenolol (7.32 ± 0.60 × 10-7 cm·s-1; mean ± SEM, n = 3-6), while the end-point amount of acamprosate internalized by the cell monolayer, Qmonolayer, was lower than that of mannitol. Acamprosate did not influence the barrier function of the monolayers since it altered neither the Papp of the three paracellular markers nor the transepithelial electrical resistance (TEER) of the cell monolayer. The Pmodeled for all the paracellular markers and acamprosate was dominated by the Ppara component and matched the experimentally obtained Papp. Furthermore, acamprosate did not inhibit the uptake of probe substrates for solute carriers PEPT1, TAUT, PAT1, EAAT1, B0,+AT/rBAT, OATP2B1, and ASBT expressed in Caco-2 cells. Thus, the Pmodeled estimated well Ppara, and the paracellular route appears to be the predominant mechanism for acamprosate Papp across Caco-2 monolayers, while the alternative transcellular routes, mediated by passive diffusion or carriers, are suggested to only play insignificant roles.

Place, publisher, year, edition, pages
American Chemical Society (ACS), 2019
Keywords
Caco-2, acamprosate, paracellular modeling, paracellular permeability, permeability modeling, permeability-limited absorption, transcellular permeability
National Category
Pharmacology and Toxicology Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-395488 (URN)10.1021/acs.molpharmaceut.9b00733 (DOI)000494894300019 ()31560549 (PubMedID)
Funder
NordForsk
Available from: 2019-10-19 Created: 2019-10-19 Last updated: 2019-11-26Bibliographically approved
Vallianatou, T., Strittmatter, N., Nilsson, A., Shariatgorji, M., Hamm, G., Pereira, M., . . . Andrén, P. E. (2018). A mass spectrometry imaging approach for investigating how drug-drug interactions influence drug blood-brain barrier permeability. NeuroImage, 172, 808-816
Open this publication in new window or tab >>A mass spectrometry imaging approach for investigating how drug-drug interactions influence drug blood-brain barrier permeability
Show others...
2018 (English)In: NeuroImage, ISSN 1053-8119, E-ISSN 1095-9572, Vol. 172, p. 808-816Article in journal (Refereed) Published
Abstract [en]

There is a high need to develop quantitative imaging methods capable of providing detailed brain localization information of several molecular species simultaneously. In addition, extensive information on the effect of the blood-brain barrier on the penetration, distribution and efficacy of neuroactive compounds is required. Thus, we have developed a mass spectrometry imaging method to visualize and quantify the brain distribution of drugs with varying blood-brain barrier permeability. With this approach, we were able to determine blood-brain barrier transport of different drugs and define the drug distribution in very small brain structures (e.g., choroid plexus) due to the high spatial resolution provided. Simultaneously, we investigated the effect of drug-drug interactions by inhibiting the membrane transporter multidrug resistance 1 protein. We propose that the described approach can serve as a valuable analytical tool during the development of neuroactive drugs, as it can provide physiologically relevant information often neglected by traditional imaging technologies.

Keywords
Mass spectrometry imaging, Blood-brain barrier, Drug-drug interactions, Elacridar, Loperamide, Propranolol
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-353358 (URN)10.1016/j.neuroimage.2018.01.013 (DOI)000430364100067 ()29329980 (PubMedID)
Funder
Swedish Research Council, 2013-3105]Swedish Research Council, 2014-6215]Swedish Foundation for Strategic Research , RIF14-0078]AstraZenecaEU, FP7, Seventh Framework Programme, 607517The Swedish Brain FoundationScience for Life Laboratory - a national resource center for high-throughput molecular bioscience
Available from: 2018-06-12 Created: 2018-06-12 Last updated: 2019-09-02Bibliographically approved
Skogh, A., Lesniak, A., Sköld, C., Karlgren, M., Gaugaz, F. Z., Svensson, R., . . . Johansson, A. (2018). An imidazole based H-Phe-Phe-NH2 peptidomimetic with anti-allodynic effect in spared nerve injury mice. Bioorganic & Medicinal Chemistry Letters, 28(14), 2446-2450
Open this publication in new window or tab >>An imidazole based H-Phe-Phe-NH2 peptidomimetic with anti-allodynic effect in spared nerve injury mice
Show others...
2018 (English)In: Bioorganic & Medicinal Chemistry Letters, ISSN 0960-894X, E-ISSN 1464-3405, Vol. 28, no 14, p. 2446-2450Article in journal (Refereed) Published
Abstract [en]

The dipeptide amide H-Phe-Phe-NH2 (1) that previously was identified as a ligand for the substance P 1-7 (SP1-7) binding site exerts intriguing results in animal models of neuropathic pain after central but not after peripheral administration. The dipeptide 1 is derived from stepwise modifications of the anti-nociceptive heptapeptide SP1-7 and the tetrapeptide endomorphin-2 that is also binding to the SP1-7 site. We herein report a strong anti-allodynic effect of a new H-Phe-Phe-NH2 peptidomimetic (4) comprising an imidazole ring as a bioisosteric element, in the spare nerve injury (SNI) mice model after peripheral administration. Peptidomimetic 4 was stable in plasma, displayed a fair membrane permeability and a favorable neurotoxic profile. Moreover, the effective dose (ED50) of 4 was superior as compared to gabapentin and morphine that are used in clinic.

National Category
Medicinal Chemistry
Identifiers
urn:nbn:se:uu:diva-343682 (URN)10.1016/j.bmcl.2018.06.009 (DOI)000438467200020 ()29929882 (PubMedID)
Funder
Swedish Research Council, 9459
Note

Title in dissertation reference list: An Imidazole-Based H-Phe-Phe-NH2 Peptidomimetic with Anti-Allodynic Effect in Spared Nerve Injury Mice and without Neurotoxic Liability

Available from: 2018-02-28 Created: 2018-02-28 Last updated: 2022-01-29Bibliographically approved
Karlgren, M., Simoff, I., Keiser, M., Oswald, S. & Artursson, P. (2018). CRISPR-Cas9: A New Addition to the Drug Metabolism and Disposition Tool Box. Drug Metabolism And Disposition, 46(11), 1776-1786
Open this publication in new window or tab >>CRISPR-Cas9: A New Addition to the Drug Metabolism and Disposition Tool Box
Show others...
2018 (English)In: Drug Metabolism And Disposition, ISSN 0090-9556, E-ISSN 1521-009X, Vol. 46, no 11, p. 1776-1786Article, review/survey (Refereed) Published
Abstract [en]

Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 9 (Cas9), i.e., CRISPR-Cas9, has been extensively used as a gene-editing technology during recent years. Unlike earlier technologies for gene editing or gene knockdown, such as zinc finger nucleases and RNA interference, CRISPR-Cas9 is comparably easy to use, affordable, and versatile. Recently, CRISPR-Cas9 has been applied in studies of drug absorption, distribution, metabolism, and excretion (ADME) and for ADME model generation. To date, about 50 papers have been published describing in vitro or in vivo CRISPR-Cas9 gene editing of ADME and ADME-related genes. Twenty of these papers describe gene editing of clinically relevant genes, such as ATP-binding cassette drug transporters and cytochrome P450 drug-metabolizing enzymes. With CRISPR-Cas9, the ADME tool box has been substantially expanded. This new technology allows us to develop better and more predictive in vitro and in vivo ADME models and map previously underexplored ADME genes and gene families. In this mini-review, we give an overview of the CRISPR-Cas9 technology and summarize recent applications of CRISPR-Cas9 within the ADME field. We also speculate about future applications of CRISPR-Cas9 in ADME research.

Place, publisher, year, edition, pages
The American Society for Pharmacology and Experimental Therapeutics, 2018
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-386345 (URN)10.1124/dmd.118.082842 (DOI)000452484200034 ()30126863 (PubMedID)
Funder
Swedish Research Council, 1951
Available from: 2019-10-16 Created: 2019-10-16 Last updated: 2019-10-16Bibliographically approved
Projects
Development of permeability enhancers for oral peptide drug delivery [2023-02733_VR]; Uppsala UniversityA miniaturized gut for optimization of oral medicines [2023-02916_VR]; Uppsala University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-4500-0170

Search in DiVA

Show all publications