Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 31(6): 793-794, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848685

ABSTRACT

Getting mature and functional stem cell-derived, insulin-producing ß cells is an important step for disease modeling, drug screening, and cell replacement therapy. In this issue, Hua et al.1 used single-cell multiomics analysis coupled with chemical screening to identify a crucial role for ceramides in generating mature stem cell-derived ß cells.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Humans , Animals , Stem Cells/cytology , Stem Cells/metabolism , Ceramides/metabolism , Single-Cell Analysis
2.
Nat Nanotechnol ; 19(2): 237-245, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37813939

ABSTRACT

Insulin binds the insulin receptor (IR) and regulates anabolic processes in target tissues. Impaired IR signalling is associated with multiple diseases, including diabetes, cancer and neurodegenerative disorders. IRs have been reported to form nanoclusters at the cell membrane in several cell types, even in the absence of insulin binding. Here we exploit the nanoscale spatial organization of the IR to achieve controlled multivalent receptor activation. To control insulin nanoscale spatial organization and valency, we developed rod-like insulin-DNA origami nanostructures carrying different numbers of insulin molecules with defined spacings. Increasing the insulin valency per nanostructure markedly extended the residence time of insulin-DNA origami nanostructures at the receptors. Both insulin valency and spacing affected the levels of IR activation in adipocytes. Moreover, the multivalent insulin design associated with the highest levels of IR activation also induced insulin-mediated transcriptional responses more effectively than the corresponding monovalent insulin nanostructures. In an in vivo zebrafish model of diabetes, treatment with multivalent-but not monovalent-insulin nanostructures elicited a reduction in glucose levels. Our results show that the control of insulin multivalency and spatial organization with nanoscale precision modulates the IR responses, independent of the insulin concentration. Therefore, we propose insulin nanoscale organization as a design parameter in developing new insulin therapies.


Subject(s)
DNA , Nanostructures , Receptor, Insulin , Animals , Diabetes Mellitus/drug therapy , DNA/chemistry , Insulin , Nanostructures/chemistry , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Zebrafish
3.
Dev Cell ; 58(6): 450-460.e6, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36893754

ABSTRACT

Building a blastema from the stump is a key step of salamander limb regeneration. Stump-derived cells temporarily suspend their identity as they contribute to the blastema by a process generally referred to as dedifferentiation. Here, we provide evidence for a mechanism that involves an active inhibition of protein synthesis during blastema formation and growth. Relieving this inhibition results in a higher number of cycling cells and enhances the pace of limb regeneration. By small RNA profiling and fate mapping of skeletal muscle progeny as a cellular model for dedifferentiation, we find that the downregulation of miR-10b-5p is critical for rebooting the translation machinery. miR-10b-5p targets ribosomal mRNAs, and its artificial upregulation causes decreased blastema cell proliferation, reduction in transcripts that encode ribosomal subunits, diminished nascent protein synthesis, and retardation of limb regeneration. Taken together, our data identify a link between miRNA regulation, ribosome biogenesis, and protein synthesis during newt limb regeneration.


Subject(s)
MicroRNAs , RNA, Small Untranslated , Animals , Urodela/genetics , RNA, Small Untranslated/metabolism , Muscle, Skeletal/metabolism , Ribosomes/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Extremities/physiology
4.
PLoS One ; 17(8): e0272046, 2022.
Article in English | MEDLINE | ID: mdl-35951607

ABSTRACT

INTRODUCTION: Inconsistent results have been reported on the association between folic acid use in pregnancy and risk of GDM. The aim of this study was to estimate the association between folic acid use and GDM in two population-based Nordic cohorts. MATERIAL AND METHODS: Two cohort studies were conducted using data from the national population registers in Norway (2005-2018, n = 791,709) and Sweden (2006-2016, n = 1,112,817). Logistic regression was used to estimate the associations between GDM and self-reported folic acid use and prescribed folic acid use, compared to non-users, adjusting for covariates. To quantify how potential unmeasured confounders may affect the estimates, E-values were reported. An exposure misclassification bias analysis was also performed. RESULTS: In Norwegian and Swedish cohorts, adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for maternal self-reported folic acid use were 1.10 (1.06-1.14) and 0.89 (0.85-0.93), with E-values of 1.43 (1.31) and 1.50 (1.36), respectively. For prescribed folic acid use, ORs were 1.33 (1.15-1.53) and 1.56 (1.41-1.74), with E-values of 1.99 (1.57) and 2.49 (2.17), in Norway and Sweden respectively. CONCLUSIONS: The slightly higher or lower odds for GDM in self-reported users of folic acid in Norway and Sweden respectively, are likely not of clinical relevance and recommendations for folic acid use in pregnancy should remain unchanged. The two Nordic cohorts showed different directions of the association between self-reported folic acid use and GDM, but based on bias analysis, exposure misclassification is an unlikely explanation since there may still be differences in prevalence of use and residual confounding. Prescribed folic acid is used by women with specific comorbidities and co-medications, which likely underlies the higher odds for GDM.


Subject(s)
Diabetes, Gestational , Cohort Studies , Diabetes, Gestational/epidemiology , Female , Folic Acid/therapeutic use , Humans , Logistic Models , Odds Ratio , Pregnancy
5.
Nat Chem Biol ; 18(9): 942-953, 2022 09.
Article in English | MEDLINE | ID: mdl-35697798

ABSTRACT

Regenerating pancreatic ß-cells is a potential curative approach for diabetes. We previously identified the small molecule CID661578 as a potent inducer of ß-cell regeneration, but its target and mechanism of action have remained unknown. We now screened 257 million yeast clones and determined that CID661578 targets MAP kinase-interacting serine/threonine kinase 2 (MNK2), an interaction we genetically validated in vivo. CID661578 increased ß-cell neogenesis from ductal cells in zebrafish, neonatal pig islet aggregates and human pancreatic ductal organoids. Mechanistically, we found that CID661578 boosts protein synthesis and regeneration by blocking MNK2 from binding eIF4G in the translation initiation complex at the mRNA cap. Unexpectedly, this blocking activity augmented eIF4E phosphorylation depending on MNK1 and bolstered the interaction between eIF4E and eIF4G, which is necessary for both hypertranslation and ß-cell regeneration. Taken together, our findings demonstrate a targetable role of MNK2-controlled translation in ß-cell regeneration, a role that warrants further investigation in diabetes.


Subject(s)
Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Animals , Cell Line , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Humans , Infant, Newborn , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Zebrafish/metabolism
6.
Pediatr Diabetes ; 22(7): 969-973, 2021 11.
Article in English | MEDLINE | ID: mdl-34487407

ABSTRACT

BACKGROUND: Experimental animal studies suggest a novel role for the folate receptor 1 in ß-cell differentiation in the pancreas, with potential implications for glycemic control. We tested the hypothesis of a protective association between prenatal folic acid use and neonatal diabetes or hyperglycemia and type 1 diabetes in an observational cohort study using data from the national population health registers in Norway. METHODS: All singleton pregnancies resulting in live births from 2005 to 2018 were identified. Prenatal exposure to folic acid was determined based on maternal report at antenatal care in early pregnancy. Diagnoses of neonatal diabetes, hyperglycemia, and type 1 diabetes for the children were identified. Associations were estimated with logistic regression or Cox proportional hazard model and included crude and adjusted estimates. RESULTS: Among 781,567 children, 69% had prenatal exposure to folic acid, 264 were diagnosed with neonatal diabetes or hyperglycemia, and 1390 with type 1 diabetes. Compared to children with no prenatal exposure to folic acid, children with prenatal exposure to folic acid had similar odds of having a neonatal diabetes or hyperglycemia diagnosis (adjusted odds ratio 0.95, 95% confidence interval [CI] 0.72, 1.25) and similar risk of being diagnosed with type 1 diabetes (adjusted hazard ratio 1.05, 95% CI 0.93, 1.18). CONCLUSIONS: No association between prenatal folic acid exposure and neonatal diabetes/hyperglycemia or type 1 diabetes was found. These findings do not rule out a translational effect of the experimental results and future studies with longer follow-up and more precise information on the window of prenatal exposure are needed.


Subject(s)
Diabetes Mellitus, Type 1/epidemiology , Folic Acid/administration & dosage , Hyperglycemia/epidemiology , Infant, Newborn, Diseases/epidemiology , Adult , Body Mass Index , Cohort Studies , Educational Status , Female , Follow-Up Studies , Humans , Infant, Newborn , Maternal-Fetal Exchange , Middle Aged , Norway/epidemiology , Pregnancy , Registries , Risk Factors , Smoking/epidemiology
7.
Nat Commun ; 12(1): 3362, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099692

ABSTRACT

Diabetes can be caused by an insufficiency in ß-cell mass. Here, we performed a genetic screen in a zebrafish model of ß-cell loss to identify pathways promoting ß-cell regeneration. We found that both folate receptor 1 (folr1) overexpression and treatment with folinic acid, stimulated ß-cell differentiation in zebrafish. Treatment with folinic acid also stimulated ß-cell differentiation in cultures of neonatal pig islets, showing that the effect could be translated to a mammalian system. In both zebrafish and neonatal pig islets, the increased ß-cell differentiation originated from ductal cells. Mechanistically, comparative metabolomic analysis of zebrafish with/without ß-cell ablation and with/without folinic acid treatment indicated ß-cell regeneration could be attributed to changes in the pyrimidine, carnitine, and serine pathways. Overall, our results suggest evolutionarily conserved and previously unknown roles for folic acid and one-carbon metabolism in the generation of ß-cells.


Subject(s)
Carbon/metabolism , Cell Differentiation/drug effects , Folate Receptor 1/metabolism , Insulin-Secreting Cells/metabolism , Leucovorin/pharmacology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Carnitine/metabolism , Cell Differentiation/genetics , Cells, Cultured , Folate Receptor 1/genetics , Gene Expression Regulation/drug effects , Humans , Insulin-Secreting Cells/cytology , Larva/genetics , Larva/metabolism , Metabolic Networks and Pathways/drug effects , Mice , Pyrimidines/metabolism , Swine , Zebrafish/genetics
8.
EMBO J ; 35(18): 2026-44, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27516442

ABSTRACT

There is great interest in therapeutically harnessing endogenous regenerative mechanisms to increase the number of ß cells in people with diabetes. By performing whole-genome expression profiling of zebrafish islets, we identified 11 secreted proteins that are upregulated during ß-cell regeneration. We then tested the proteins' ability to potentiate ß-cell regeneration in zebrafish at supraphysiological levels. One protein, insulin-like growth factor (Igf) binding-protein 1 (Igfbp1), potently promoted ß-cell regeneration by potentiating α- to ß-cell transdifferentiation. Using various inhibitors and activators of the Igf pathway, we show that Igfbp1 exerts its regenerative effect, at least partly, by inhibiting Igf signaling. Igfbp1's effect on transdifferentiation appears conserved across species: Treating mouse and human islets with recombinant IGFBP1 in vitro increased the number of cells co-expressing insulin and glucagon threefold. Moreover, a prospective human study showed that having high IGFBP1 levels reduces the risk of developing type-2 diabetes by more than 85%. Thus, we identify IGFBP1 as an endogenous promoter of ß-cell regeneration and highlight its clinical importance in diabetes.


Subject(s)
Cell Transdifferentiation , Glucagon-Secreting Cells/physiology , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Secreting Cells/physiology , Regeneration , Animals , Humans , Mice , Zebrafish
9.
Cell Metab ; 23(1): 194-205, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26701651

ABSTRACT

Although compensatory islet hyperplasia in response to insulin resistance is a recognized feature in diabetes, the factor(s) that promote ß cell proliferation have been elusive. We previously reported that the liver is a source for such factors in the liver insulin receptor knockout (LIRKO) mouse, an insulin resistance model that manifests islet hyperplasia. Using proteomics we show that serpinB1, a protease inhibitor, which is abundant in the hepatocyte secretome and sera derived from LIRKO mice, is the liver-derived secretory protein that regulates ß cell proliferation in humans, mice, and zebrafish. Small-molecule compounds, that partially mimic serpinB1 effects of inhibiting elastase activity, enhanced proliferation of ß cells, and mice lacking serpinB1 exhibit attenuated ß cell compensation in response to insulin resistance. Finally, SerpinB1 treatment of islets modulated proteins in growth/survival pathways. Together, these data implicate serpinB1 as an endogenous protein that can potentially be harnessed to enhance functional ß cell mass in patients with diabetes.


Subject(s)
Cell Proliferation , Insulin-Secreting Cells/physiology , Serpins/physiology , Animals , Cells, Cultured , Humans , Insulin Resistance , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...