Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
STAR Protoc ; 4(1): 102008, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36640368

ABSTRACT

The ability of stem cells to activate and differentiate is critical for maintaining the regenerative capacity of skeletal muscle. Here, we detail steps for specific quantification and isolation of primary human fibro-adipogenic progenitors and skeletal muscle stem cells using fluorescence-activated cell sorting. We describe important phenotypic traits such as time to enter the cell cycle and assessment of cell differentiation for the isolated cell populations. The technique has been applied on tissue obtained from surgery and needle biopsies. For complete details on the use and execution of this protocol, please refer to Farup et al. (2021).1.


Subject(s)
Adipogenesis , Stem Cells , Humans , Flow Cytometry/methods , Cell Differentiation/physiology , Muscle, Skeletal
2.
JCI Insight ; 7(19)2022 10 10.
Article in English | MEDLINE | ID: mdl-35998039

ABSTRACT

BACKGROUNDDuring aging, there is a functional decline in the pool of muscle stem cells (MuSCs) that influences the functional and regenerative capacity of skeletal muscle. Preclinical evidence has suggested that nicotinamide riboside (NR) and pterostilbene (PT) can improve muscle regeneration, e.g., by increasing MuSC function. The objective of this study was to investigate if supplementation with NR and PT (NRPT) promotes skeletal muscle regeneration after muscle injury in elderly individuals by improved recruitment of MuSCs.METHODSThirty-two elderly individuals (55-80 years of age) were randomized to daily supplementation with either NRPT (1,000 mg NR and 200 mg PT) or matched placebo. Two weeks after initiation of supplementation, skeletal muscle injury was induced by electrically induced eccentric muscle work. Skeletal muscle biopsies were obtained before, 2 hours after, and 2, 8, and 30 days after injury.RESULTSA substantial skeletal muscle injury was induced by the protocol and associated with release of myoglobin and creatine kinase, muscle soreness, tissue edema, and a decrease in muscle strength. MuSC content, proliferation, and cell size revealed a large demand for recruitment after injury, but this was not affected by NRPT. Furthermore, histological analyses of muscle fiber area, central nuclei, and embryonic myosin heavy chain showed no NRPT supplementation effect.CONCLUSIONDaily supplementation with 1,000 mg NR and 200 mg PT is safe but does not improve recruitment of the MuSC pool or other measures of muscle recovery in response to injury or subsequent regeneration in elderly individuals.TRIAL REGISTRATIONClinicalTrials.gov NCT03754842.FUNDINGNovo Nordisk Foundation (NNF17OC0027242) and Novo Nordisk Foundation CBMR.


Subject(s)
Muscular Diseases , Myosin Heavy Chains , Aged , Creatine Kinase, MM Form , Dietary Supplements , Humans , Muscle, Skeletal , Myoglobin/pharmacology , Niacinamide/analogs & derivatives , Pyridinium Compounds , Stilbenes
3.
Cell Metab ; 33(11): 2201-2214.e11, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34678202

ABSTRACT

Type 2 diabetes mellitus (T2DM) is associated with impaired skeletal muscle function and degeneration of the skeletal muscles. However, the mechanisms underlying the degeneration are not well described in human skeletal muscle. Here we show that skeletal muscle of T2DM patients exhibit degenerative remodeling of the extracellular matrix that is associated with a selective increase of a subpopulation of fibro-adipogenic progenitors (FAPs) marked by expression of THY1 (CD90)-the FAPCD90+. We identify platelet-derived growth factor (PDGF) as a key FAP regulator, as it promotes proliferation and collagen production at the expense of adipogenesis. FAPsCD90+ display a PDGF-mimetic phenotype, with high proliferative activity, clonogenicity, and production of extracellular matrix. FAPCD90+ proliferation was reduced by in vitro treatment with metformin. Furthermore, metformin treatment reduced FAP content in T2DM patients. These data identify a PDGF-driven conversion of a subpopulation of FAPs as a key event in the fibrosis development in T2DM muscle.


Subject(s)
Diabetes Mellitus, Type 2 , Muscular Diseases , Adipogenesis , Cell Differentiation , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Humans , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Diseases/metabolism
4.
Mol Ther Nucleic Acids ; 24: 403-415, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-33868784

ABSTRACT

CRISPR gene therapy is one promising approach for treatment of Duchenne muscular dystrophy (DMD), which is caused by a large spectrum of mutations in the dystrophin gene. To broaden CRISPR gene editing strategies for DMD treatment, we report the efficient restoration of dystrophin expression in induced myotubes by SpCas9 and dual guide RNAs (gRNAs). We first sequenced 32 deletion junctions generated by this editing method and revealed that non-homologous blunt-end joining represents the major indel type. Based on this predictive repair outcome, efficient in-frame deletion of a part of DMD exon 51 was achieved in HEK293T cells with plasmids expressing SpCas9 and dual gRNAs. More importantly, we further corrected a frameshift mutation in human DMD (exon45del) fibroblasts with SpCas9-dual gRNA ribonucleoproteins. The edited DMD fibroblasts were transdifferentiated into myotubes by lentiviral-mediated overexpression of a human MYOD transcription factor. Restoration of DMD expression at both the mRNA and protein levels was confirmed in the induced myotubes. With further development, the combination of SpCas9-dual gRNA-corrected DMD patient fibroblasts and transdifferentiation may provide a valuable therapeutic strategy for DMD.

5.
J Clin Endocrinol Metab ; 105(11)2020 11 01.
Article in English | MEDLINE | ID: mdl-32480406

ABSTRACT

Metformin has undisputed glucose-lowering effects in diabetes and an impressive safety record. It has also shown promising effects beyond diabetes, and several hundred clinical trials involving metformin are currently planned or active. Metformin targets intracellular effectors, but exactly which remain to be established, and in an era of precision medicine, an incomplete understanding of mechanisms of action may limit the use of metformin. Distribution of metformin depends on specific organic cation transporter proteins that are organ- and species-specific. Therefore, target tissues of metformin can be identified by cellular uptake of the drug, and exploring the biodistribution of the drug in humans becomes an attractive strategy to assist the many investigations into the mechanisms of action of metformin performed in animals. In this review, we combine the emerging evidence from the use of 11C-labeled metformin in humans to discuss metformin action in liver, intestines, and kidney, which are the organs with the most avid uptake of the drug.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Diabetes Mellitus, Type 2/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Tissue Distribution
6.
J Nucl Med ; 57(12): 1920-1926, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27469359

ABSTRACT

Metformin is the most widely prescribed oral antiglycemic drug, with few adverse effects. However, surprisingly little is known about its human biodistribution and target tissue metabolism. In animal experiments, we have shown that metformin can be labeled by 11C and that 11C-metformin PET can be used to measure renal function. Here, we extend these preclinical findings by a first-in-human 11C-metformin PET dosimetry, biodistribution, and tissue kinetics study. METHODS: Nine subjects (3 women and 6 men) participated in 2 studies: in the first study, human radiation dosimetry and biodistribution of 11C-metformin were estimated in 4 subjects (2 women and 2 men) by whole-body PET. In the second study, 11C-metformin tissue kinetics were measured in response to both intravenous and oral radiotracer administration. A dynamic PET scan with a field of view covering target tissues of metformin (liver, kidneys, intestines, and skeletal muscle) was obtained for 90 (intravenous) and 120 (oral) min. RESULTS: Radiation dosimetry was acceptable, with effective doses of 9.5 µSv/MBq (intravenous administration) and 18.1 µSv/MBq (oral administration). Whole-body PET revealed that 11C-metformin was primarily taken up by the kidneys, urinary bladder, and liver but also to a lesser extent in salivary glands, skeletal muscle, and intestines. Reversible 2-tissue-compartment kinetics was observed in the liver, and volume of distribution was calculated to be 2.45 mL/mL (arterial input) or 2.66 mL/mL (portal and arterial input). In the kidneys, compartmental models did not adequately fit the experimental data, and volume of distribution was therefore estimated by a linear approach to be 6.83 mL/mL. Skeletal muscle and intestinal tissue kinetics were best described by 2-tissue-compartment kinetics and showed only discrete tracer uptake. Liver 11C-metformin uptake was pronounced after oral administration of the tracer, with tissue-to-blood ratio double what was observed after intravenous administration. Only slow accumulation of 11C-metformin was observed in muscle. There was no elimination of 11C-metformin through the bile both during the intravenous and during the oral part of the study. CONCLUSION: 11C-metformin is suitable for imaging metformin uptake in target tissues and may prove a valuable tool to assess the impact of metformin treatment in patients with varying metformin transport capacity.


Subject(s)
Carbon Radioisotopes , Metformin/pharmacokinetics , Positron-Emission Tomography/methods , Adult , Female , Humans , Kinetics , Male , Middle Aged , Radiometry , Tissue Distribution , Whole Body Imaging
7.
EJNMMI Res ; 6(1): 54, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27339044

ABSTRACT

BACKGROUND: Organic cation transporters (OCTs) in the renal proximal tubule are important for the excretion of both exo- and endogenous compounds, and chronic kidney disease (CKD) alter the expression of OCT. Metformin is a well-known substrate for OCT, and recently, we demonstrated that positron emission tomography (PET) with 11C-labelled metformin ((11)C-metformin) is a promising approach to evaluate the function of OCT. The aim of this study is therefore to examine renal pharmacokinetics of (11)C-metformin and expression of OCTs in a transgenic (RenTGF-ß1) mouse model of CKD. METHODS: Age- and sex-matched RenTGF-ß1 (Tg) and wildtype (WT) mice were used (5-8/group). Animals received an iv bolus of (11)C-metformin followed by 90-min dynamic PET and MRI scan. PET data were analysed using a one-tissue compartment model. Renal protein abundance of OCT2 (by Western blot) as well as OCT1, OCT2, and MATE1 messenger RNA (mRNA) (by RT-PCR) was examined. RESULTS: Protein expression of the basolateral uptake transporter OCT2 was 1.5-fold lower in Tg mice compared to WT mice while OCT1 and MATE1 mRNA expression did not differ between the two groups. The influx rate constant of (11)C-metformin in renal cortex (K 1) was 2.2-fold lower in transgenic mice whereas the backflux rate constant (k 2) was similar in the two groups, consistent with protein expression. Total body clearance (TBC) correlated within each group linearly with K 1. CONCLUSIONS: In conclusion, this study demonstrates that both renal OCT2 expression and (11)C-metformin uptake are reduced in CKD mice. This potentially makes (11)C-metformin valuable as a PET probe to evaluate kidney function.

8.
J Nucl Med ; 57(4): 615-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26769859

ABSTRACT

UNLABELLED: Organic cation transporters (OCTs) in the kidney proximal tubule (PT) participate in renal excretion of drugs and endogenous compounds. PT function is commonly impaired in kidney diseases, and consequently quantitative measurement of OCT function may provide an important estimate of kidney function. Metformin is a widely used drug and targets OCT type 2 located in the PT. Thus, we hypothesized that (11)C-labeled metformin would be a suitable PET tracer for quantification of renal function. METHODS: (11)C-metformin was prepared by (11)C-methylation of 1-methylbiguanide. In vitro cell uptake of (11)C-metformin was studied in LLC-PK1 cells in the presence of increasing doses of unlabeled metformin. In vivo small-animal PET studies in Sprague-Dawley rats were performed at baseline and after treatment with OCT inhibitors to evaluate renal uptake of (11)C-metformin. Kidney and liver pharmacokinetics of (11)C-metformin was investigated in vivo by dynamic (11)C-metformin PET/CT in 6 anesthetized pigs, and renal clearance of (11)C-metformin was compared with renal clearance of (51)Cr-ethylenediaminetetraacetic acid (EDTA). Formation of (11)C metabolites was investigated by analysis of blood and urine samples. RESULTS: The radiochemical yield of (11)C-metformin was 15% ± 3% (n= 40, decay-corrected), and up to 1.5 GBq of tracer were produced with a radiochemical purity greater than 95% in less than 30 min. Dose-dependent uptake of (11)C-metformin in LLC-PK1 cells was rapid. Rat small-animal PET images showed (11)C-metformin uptake in the kidney and liver, the kinetics of which were changed after challenging animals with OCT inhibitors. In pigs, 80% of the injected metformin dose was rapidly present in the kidney, and a high dose of metformin caused a delayed renal uptake and clearance compared with baseline consistent with transporter-mediated competition. Renal clearance of (11)C-metformin was approximately 3 times the renal clearance of (51)Cr-EDTA. CONCLUSION: We successfully synthesized an (11)C-metformin tracer, and PET studies in rats and pigs showed a rapid kidney uptake from the blood and excretion into the bladder similar to other radiopharmaceuticals developed for γ-camera renography.


Subject(s)
Kidney/diagnostic imaging , Kidney/metabolism , Metformin/chemical synthesis , Metformin/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Animals , Autoradiography , Biguanides/chemistry , Dose-Response Relationship, Drug , Edetic Acid/pharmacokinetics , Isotope Labeling , LLC-PK1 Cells , Liver/metabolism , Methylation , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Swine , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...