Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
2.
Phys Rev E ; 109(1-1): 014140, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38366424

ABSTRACT

This paper solves in one and two dimensions the steady noninteractive active Fokker-Planck (FP) equation and finds that its velocity distribution admits, under limiting cases, a dual behavior. Briefly, when the inertial relaxation time is smaller than the orientation time, the active FP equation admits a bimodal shape, whereas the inverse condition is seen to admit a Gaussian one. Once the velocity distribution functions are available, they are used to find their effect on the system's transport properties, such as its mean-square speed. In the process, a useful mathematical identity for the first kind Bessel function as a sum of bimodal exponential functions is spotted.

3.
Trends Cell Biol ; 34(3): 180-197, 2024 03.
Article in English | MEDLINE | ID: mdl-37626005

ABSTRACT

ß-Cell replacement by in situ reprogramming of non-ß-cells is a promising diabetes therapy. Following the observation that near-total ß-cell ablation in adult mice triggers the reprogramming of pancreatic α-, δ-, and γ-cells into insulin (INS)-producing cells, recent studies are delving deep into the mechanisms controlling adult α-cell identity. Systematic analyses of the α-cell transcriptome and epigenome have started to pinpoint features that could be crucial for maintaining α-cell identity. Using different transgenic and chemical approaches, significant advances have been made in reprogramming α-cells in vivo into INS-secreting cells in mice. The recent reprogramming of human α-cells in vitro is an important step forward that must now be complemented with a comprehensive molecular dissection of the mechanisms controlling α-cell identity.


Subject(s)
Glucagon-Secreting Cells , Insulin-Secreting Cells , Humans , Mice , Animals , Insulin , Glucagon
4.
J Invest Dermatol ; 144(2): 284-295.e16, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37716648

ABSTRACT

Desmosomes are dynamic complex protein structures involved in cellular adhesion. Disruption of these structures by loss-of-function variants in desmosomal genes leads to a variety of skin- and heart-related phenotypes. In this study, we report TUFT1 as a desmosome-associated protein, implicated in epidermal integrity. In two siblings with mild skin fragility, woolly hair, and mild palmoplantar keratoderma but without a cardiac phenotype, we identified a homozygous splice-site variant in the TUFT1 gene, leading to aberrant mRNA splicing and loss of TUFT1 protein. Patients' skin and keratinocytes showed acantholysis, perinuclear retraction of intermediate filaments, and reduced mechanical stress resistance. Immunolabeling and transfection studies showed that TUFT1 is positioned within the desmosome and that its location is dependent on the presence of the desmoplakin carboxy-terminal tail. A Tuft1-knockout mouse model mimicked the patients' phenotypes. Altogether, this study reveals TUFT1 as a desmosome-associated protein, whose absence causes skin fragility, woolly hair, and palmoplantar keratoderma.


Subject(s)
Hair Diseases , Keratoderma, Palmoplantar , Skin Abnormalities , Animals , Humans , Mice , Desmoplakins/genetics , Desmoplakins/metabolism , Desmosomes/metabolism , Hair/metabolism , Hair Diseases/genetics , Hair Diseases/metabolism , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/metabolism , Skin/metabolism , Skin Abnormalities/metabolism
5.
Cell Stem Cell ; 30(4): 488-497.e3, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028408

ABSTRACT

Understanding the origin of pancreatic ß cells has profound implications for regenerative therapies in diabetes. For over a century, it was widely held that adult pancreatic duct cells act as endocrine progenitors, but lineage-tracing experiments challenged this dogma. Gribben et al. recently used two existing lineage-tracing models and single-cell RNA sequencing to conclude that adult pancreatic ducts contain endocrine progenitors that differentiate to insulin-expressing ß cells at a physiologically important rate. We now offer an alternative interpretation of these experiments. Our data indicate that the two Cre lines that were used directly label adult islet somatostatin-producing ∂ cells, which precludes their use to assess whether ß cells originate from duct cells. Furthermore, many labeled ∂ cells, which have an elongated neuron-like shape, were likely misclassified as ß cells because insulin-somatostatin coimmunolocalizations were not used. We conclude that most evidence so far indicates that endocrine and exocrine lineage borders are rarely crossed in the adult pancreas.


Subject(s)
Insulin-Secreting Cells , Evidence Gaps , Cell Differentiation , Pancreas/physiology , Pancreatic Ducts , Insulin , Somatostatin
6.
Animals (Basel) ; 12(20)2022 Oct 11.
Article in English | MEDLINE | ID: mdl-36290119

ABSTRACT

Ca Rater Mallorquí is a dog breed from the Island of Mallorca (Spain) traditionally used as a hunting and ratting dog to prevent disease spread and economic losses related to rodent activities on farms. However, the census data shows a population decline that should be addressed by implementing a conservation program. The first step to implementing a conservation plan is knowing the genetic situation of the Ca Rater Mallorquí population. Therefore, we aimed to genetically characterise the breed in our study. We analysed 33 microsatellites recommended by the International Society of Animal Genetics (ISAG) in 77 samples. Data were obtained from 13 samples of Balearic, Spanish, and international dog breeds to study the genetic diversity among breeds. The population did not significantly deviate from the Hardy-Weinberg equilibrium with heterozygosity (Ho) of 0.655 and expected heterozygosity (He) of 0.685. The Wright's fixation indices, the Factorial Correspondence Analysis (FCA), a dendrogram representing Reynolds genetic distance between populations, and the pairwise FST values establish the Ca Rater Mallorquí as an independent breed distinct from the Balearic, Spanish, and international breeds.

7.
Sci Rep ; 12(1): 9033, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35641781

ABSTRACT

Improved models of experimental diabetes are needed to develop cell therapies for diabetes. Here, we introduce the B6 RIP-DTR mouse, a model of experimental diabetes in fully immunocompetent animals. These inbred mice harbor the H2b major histocompatibility complex (MHC), selectively express high affinity human diphtheria toxin receptor (DTR) in islet ß-cells, and are homozygous for the Ptprca (CD45.1) allele rather than wild-type Ptprcb (CD45.2). 100% of B6 RIP-DTR mice rapidly became diabetic after a single dose of diphtheria toxin, and this was reversed indefinitely after transplantation with islets from congenic C57BL/6 mice. By contrast, MHC-mismatched islets were rapidly rejected, and this allotransplant response was readily monitored via blood glucose and graft histology. In peripheral blood of B6 RIP-DTR with mixed hematopoietic chimerism, CD45.2 BALB/c donor blood immune cells were readily distinguished from host CD45.1 cells by flow cytometry. Reliable diabetes induction and other properties in B6 RIP-DTR mice provide an important new tool to advance transplant-based studies of islet replacement and immunomodulation to treat diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Diabetes Mellitus, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation Immunology
8.
Nat Commun ; 13(1): 2020, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440614

ABSTRACT

Generation of surrogate cells with stable functional identities is crucial for developing cell-based therapies. Efforts to produce insulin-secreting replacement cells to treat diabetes require reliable tools to assess islet cellular identity. Here, we conduct a thorough single-cell transcriptomics meta-analysis to identify robustly expressed markers used to build genesets describing the identity of human α-, ß-, γ- and δ-cells. These genesets define islet cellular identities better than previously published genesets. We show their efficacy to outline cell identity changes and unravel some of their underlying genetic mechanisms, whether during embryonic pancreas development or in experimental setups aiming at developing glucose-responsive insulin-secreting cells, such as pluripotent stem-cell differentiation or in adult islet cell reprogramming protocols. These islet cell type-specific genesets represent valuable tools that accurately benchmark gain and loss in islet cell identity traits.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Pluripotent Stem Cells , Cell Differentiation/genetics , Humans , Insulin/genetics
9.
Cell Rep ; 38(7): 110377, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172145

ABSTRACT

The precise developmental dynamics of the pancreatic islet endocrine cell types, and their interrelation, are unknown. Some authors claim the persistence of islet cell differentiation from precursor cells after birth ("neogenesis"). Here, using four conditional cell lineage tracing ("pulse-and-chase") murine models, we describe the natural history of pancreatic islet cells, once they express a hormone gene, until late in life. Concerning the contribution of early-appearing embryonic hormone-expressing cells to the formation of islets, we report that adult islet cells emerge from embryonic hormone-expressing cells arising at different time points during development, without any evidence of postnatal neogenesis. We observe specific patterns of hormone gene activation and switching during islet morphogenesis, revealing that, within each cell type, cells have heterogeneous developmental trajectories. This likely applies to most maturating cells in the body, and explains the observed phenotypic variability within differentiated cell types. Such knowledge should help devising novel regenerative therapies.


Subject(s)
Aging/physiology , Fetus/cytology , Hormones/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Animals , Doxycycline/pharmacology , Embryonic Development/drug effects , Fetus/drug effects , Gene Expression Regulation, Developmental/drug effects , Glucagon/metabolism , Islets of Langerhans/drug effects , Mice, Transgenic , Somatostatin/metabolism , Staining and Labeling
10.
Dalton Trans ; 50(46): 16939-16944, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34779455

ABSTRACT

Metal-organic Frameworks (MOFs) have emerged as promising materials for different electrochemical applications. Their low conductivity, however, is a major challenge to overcome. Therefore, a deeper understanding on the charge transfer mechanism is needed to improve the conductivity of MOF-based electrodes. For this contribution, we focused on metalated MOF-525 and found that the nature of the metal center is one of the many factors contributing to the charge transfer kinetics, which is attributed to differences in redox behaviour, affecting the hopping distance and the electron transfer rate. These results highlight the importance of the nature of the redox active site to optimize charge transfer in MOF-based electrodes.

12.
Rev. colomb. anestesiol ; 49(3): e400, July-Sept. 2021. tab
Article in English | LILACS, COLNAL | ID: biblio-1280181

ABSTRACT

Abstract Several findings paved the way to the use of the spinal approach for anesthesia. Information about the originators and dates of their discoveries is controversial. According to personal communications, doctors Juan Bautista Montoya y Flórez, in Medellín, and Lisandro Leyva, in Bogotá, in 1904 and 1905 respectively, would appear to be the pioneers of spinal anesthesia in Colombia. Pioneering cases of this procedure carried out in 1901 by doctor Juan Evangelista Manrique and which continued to be performed by his colleagues and assistants of the medical community at the time are documented in the Corporis Fabrica dissertation collection of the National University of Colombia.


Resumen Son varios los hallazgos que contribuyeron al abordaje de la columna vertebral como vía para la administración de anestesia. Los autores y las fechas de tales descubrimientos han sido controvertidos. En Colombia, según fuentes orales, los doctores Juan Bautista Montoya y Flórez, en Medellín, y Lisandro Leyva, en Bogotá, en 1904 y 1905 respectivamente, serían los pioneros de la anestesia raquídea en Colombia. En las tesis de la Facultad de Medicina de la Universidad Nacional, Colección Corporis Fabrica, se documentan los casos pioneros de este procedimiento realizados en 1901, por el doctor Juan Evangelista Manrique y continuados enseguida por sus colegas y ayudantes de la comunidad médica de ese entonces.


Subject(s)
Humans , Anesthesia , Anesthesia, Spinal/history , Anesthetics, Local , Schools, Medical , Spine , Methods
13.
BMC Genomics ; 22(1): 585, 2021 Aug 02.
Article in English | MEDLINE | ID: mdl-34340653

ABSTRACT

BACKGROUND: Loss of pancreatic insulin-secreting ß-cells due to metabolic or autoimmune damage leads to the development of diabetes. The discovery that α-cells can be efficiently reprogrammed into insulin-secreting cells in mice and humans has opened promising avenues for innovative diabetes therapies. ß-cell loss triggers spontaneous reprogramming of only 1-2% of α-cells, limiting the extent of regeneration. Most α-cells are refractory to conversion and their global transcriptomic response to severe ß-cell loss as well as the mechanisms opposing their reprogramming into insulin producers are largely unknown. Here, we performed RNA-seq on FAC-sorted α-cells to characterize their global transcriptional responses at different time points after massive ß-cell ablation. RESULTS: Our results show that α-cells undergo stage-specific transcriptional changes 5- and 15-days post-diphtheria toxin (DT)-mediated ß-cell ablation. At 5 days, α-cells transiently upregulate various genes associated with interferon signaling and proliferation, including Interferon Induced Protein with Tetratricopeptide Repeats 3 (Ifit3). Subsequently, at 15 days post ß-cell ablation, α-cells undergo a transient downregulation of genes from several pathways including Insulin receptor, mTOR and MET signaling. CONCLUSIONS: The results presented here pinpoint novel markers discriminating α-cells at different stages after acute ß-cell loss, and highlight additional signaling pathways that are modulated in α-cells in this context.


Subject(s)
Diabetes Mellitus , Glucagon-Secreting Cells , Insulin-Secreting Cells , Animals , Insulin , Mice , Transcriptome
14.
Nat Commun ; 12(1): 4458, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294685

ABSTRACT

The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by ß-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice, Ppy gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon ß-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.


Subject(s)
Insulin/biosynthesis , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreatic Polypeptide/metabolism , Protein Precursors/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Cell Lineage/genetics , Female , Gene Knock-In Techniques , Humans , Insulin-Secreting Cells/classification , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Pancreas/cytology , Pancreas/embryology , Pancreas/growth & development , Pancreatic Polypeptide/deficiency , Pancreatic Polypeptide/genetics , Pancreatic Polypeptide-Secreting Cells/classification , Pancreatic Polypeptide-Secreting Cells/cytology , Pregnancy , RNA-Seq
15.
J Clin Invest ; 2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34292882

ABSTRACT

The start codon c.1A>G mutation in KLHL24, encoding ubiquitin-ligase KLHL24, results in the loss of 28 N-terminal amino acids (KLHL24-ΔN28) by skipping the initial start codon. In skin, KLHL24-ΔN28 leads to gain of function, excessively targeting intermediate filament keratin-14 for proteasomal degradation, ultimately causing epidermolysis bullosa simplex (EBS). The majority of these EBS-patients are also diagnosed with dilated cardiomyopathy (DCM), but the pathological mechanism in the heart is unknown. As desmin is the cardiac homologue of keratin-14, we hypothesized that KLHL24-ΔN28 leads to excessive degradation of desmin, resulting in DCM. Dynamically loaded engineered heart tissues (dyn-EHTs) were generated from human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes from two patients and three (non)familial controls. Ten-fold lower desmin protein levels were observed in patient-derived dyn-EHTs, in line with diminished desmin levels detected in patients' explanted heart. This was accompanied by tissue dilatation, impaired mitochondrial function, decreased force values and increased cardiomyocyte stress. HEK293 transfection studies confirmed KLHL24-mediated desmin degradation. KLHL24 RNA interference or direct desmin overexpression recovered desmin protein levels, restoring morphology and function in patient-derived dyn-EHTs. To conclude, presence of KLHL24-ΔN28 in cardiomyocytes leads to excessive degradation of desmin, affecting tissue morphology and function, that can be prevented by restoring desmin protein levels.

16.
Mol Metab ; 53: 101268, 2021 11.
Article in English | MEDLINE | ID: mdl-34118477

ABSTRACT

OBJECTIVE: Glucagon is secreted by pancreatic α-cells in response to hypoglycemia and its hyperglycemic effect helps to restore normal blood glucose. Insulin and somatostatin (SST) secretions from ß- and δ-cells, respectively, are stimulated by glucose by mechanisms involving an inhibition of their ATP-sensitive K+ (KATP) channels, leading to an increase in [Ca2+]c that triggers exocytosis. Drugs that close KATP channels, such as sulfonylureas, are used to stimulate insulin release in type 2 diabetic patients. α-cells also express KATP channels. However, the mechanisms by which sulfonylureas control glucagon secretion are still largely debated and were addressed in the present study. In particular, we studied the effects of KATP channel blockers on α-cell [Ca2+]c and glucagon secretion in the presence of a low (1 mM) or a high (15 mM) glucose concentration and evaluated the role of SST in these effects. METHODS: Using a transgenic mouse model expressing the Ca2+-sensitive fluorescent protein, GCaMP6f, specifically in α-cells, we measured [Ca2+]c in α-cells either dispersed or within whole islets (by confocal microscopy). By measuring [Ca2+]c in α-cells within islets and glucagon secretion using the same perifusion protocols, we tested whether glucagon secretion correlated with changes in [Ca2+]c in response to sulfonylureas. We studied the role of SST in the effects of sulfonylureas using multiple approaches including genetic ablation of SST, or application of SST-14 and SST receptor antagonists. RESULTS: Application of the sulfonylureas, tolbutamide, or gliclazide, to a medium containing 1 mM or 15 mM glucose increased [Ca2+]c in α-cells by a direct effect as in ß-cells. At low glucose, sulfonylureas inhibited glucagon secretion of islets despite the rise in α-cell [Ca2+]c that they triggered. This glucagonostatic effect was indirect and attributed to SST because, in the islets of SST-knockout mice, sulfonylureas induced a stimulation of glucagon secretion which correlated with an increase in α-cell [Ca2+]c. Experiments with exogenous SST-14 and SST receptor antagonists indicated that the glucagonostatic effect of sulfonylureas mainly resulted from an inhibition of the efficacy of cytosolic Ca2+ on exocytosis. Although SST-14 was also able to inhibit glucagon secretion by decreasing α-cell [Ca2+]c, no decrease in [Ca2+]c occurred during sulfonylurea application because it was largely counterbalanced by the direct stimulatory effect of these drugs on α-cell [Ca2+]c. At high glucose, i.e., in conditions where glucagon release was already low, sulfonylureas stimulated glucagon secretion because their direct stimulatory effect on α-cells exceeded the indirect effect by SST. Our results also indicated that, unexpectedly, SST-14 poorly decreased the efficacy of Ca2+ on exocytosis in ß-cells. CONCLUSIONS: Sulfonylureas exert two opposite actions on α-cells: a direct stimulation as in ß-cells and an indirect inhibition by SST. This suggests that any alteration of SST paracrine influence, as described in diabetes, will modify the effect of sulfonylureas on glucagon release. In addition, we suggest that δ-cells inhibit α-cells more efficiently than ß-cells.


Subject(s)
Calcium/metabolism , Gliclazide/pharmacology , Glucagon-Secreting Cells/drug effects , Glucagon/metabolism , KATP Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Somatostatin/pharmacology , Tolbutamide/pharmacology , Animals , Gliclazide/chemistry , Glucagon-Secreting Cells/metabolism , KATP Channels/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Potassium Channel Blockers/chemistry , Somatostatin/chemistry , Tolbutamide/chemistry
17.
Sci Adv ; 7(18)2021 04.
Article in English | MEDLINE | ID: mdl-33910906

ABSTRACT

Vision is usually assumed to be sensitive to the light intensity and spectrum but not to its spectral phase. However, experiments performed on retinal proteins in solution showed that the first step of vision consists in an ultrafast photoisomerization that can be coherently controlled by shaping the phase of femtosecond laser pulses, especially in the multiphoton interaction regime. The link between these experiments in solution and the biological process allowing vision was not demonstrated. Here, we measure the electric signals fired from the retina of living mice upon femtosecond multipulse and single-pulse light stimulation. Our results show that the electrophysiological signaling is sensitive to the manipulation of the light excitation on a femtosecond time scale. The mechanism relies on multiple interactions with the light pulses close to the conical intersection, like pump-dump (photoisomerization interruption) and pump-repump (reverse isomerization) processes. This interpretation is supported both experimentally and by dynamics simulations.


Subject(s)
Light , Animals , Mice
18.
Phys Rev E ; 103(1-1): 012601, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33601560

ABSTRACT

We theoretically and computationally find a Maxwell-Boltzmann-like velocity distribution for noninteracting active matter (NAM). To achieve this, mass and moment of inertia are incorporated into the corresponding noninteracting active Fokker-Planck equation (NAFP), thus solving for the first time, the underdamped scenario of NAM following a Fokker-Planck formalism. This time, the distribution results in a bimodal symmetric expression that contains the effect of inertia on transport properties of NAM. The analytical distribution is further compared to experiments dealing with vibrobots. A generalization of the Brinkman hierarchy for NAFP is also provided and used for systematically solving the NAFP in position space. This work is an important step toward characterizing active matter using an equivalent nonequilibrium statistical mechanics.

19.
Mol Metab ; 42: 101071, 2020 12.
Article in English | MEDLINE | ID: mdl-32896668

ABSTRACT

OBJECTIVE: Sodium-glucose cotransporter 2 (SGLT2) inhibitors (SGLT2i), or gliflozins, are anti-diabetic drugs that lower glycemia by promoting glucosuria, but they also stimulate endogenous glucose and ketone body production. The likely causes of these metabolic responses are increased blood glucagon levels, and decreased blood insulin levels, but the mechanisms involved are hotly debated. This study verified whether or not SGLT2i affect glucagon and insulin secretion by a direct action on islet cells in three species, using multiple approaches. METHODS: We tested the in vivo effects of two selective SGLT2i (dapagliflozin, empagliflozin) and a SGLT1/2i (sotagliflozin) on various biological parameters (glucosuria, glycemia, glucagonemia, insulinemia) in mice. mRNA expression of SGLT2 and other glucose transporters was assessed in rat, mouse, and human FACS-purified α- and ß-cells, and by analysis of two human islet cell transcriptomic datasets. Immunodetection of SGLT2 in pancreatic tissues was performed with a validated antibody. The effects of dapagliflozin, empagliflozin, and sotagliflozin on glucagon and insulin secretion were assessed using isolated rat, mouse and human islets and the in situ perfused mouse pancreas. Finally, we tested the long-term effect of SGLT2i on glucagon gene expression. RESULTS: SGLT2 inhibition in mice increased the plasma glucagon/insulin ratio in the fasted state, an effect correlated with a decline in glycemia. Gene expression analyses and immunodetections showed no SGLT2 mRNA or protein expression in rodent and human islet cells, but moderate SGLT1 mRNA expression in human α-cells. However, functional experiments on rat, mouse, and human (29 donors) islets and the in situ perfused mouse pancreas did not identify any direct effect of dapagliflozin, empagliflozin or sotagliflozin on glucagon and insulin secretion. SGLT2i did not affect glucagon gene expression in rat and human islets. CONCLUSIONS: The data indicate that the SGLT2i-induced increase of the plasma glucagon/insulin ratio in vivo does not result from a direct action of the gliflozins on islet cells.


Subject(s)
Glucagon/metabolism , Insulin Secretion/physiology , Sodium-Glucose Transporter 2/metabolism , Animals , Benzhydryl Compounds/pharmacology , Blood Glucose/metabolism , Glucagon/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Glucosides/pharmacology , Humans , Insulin/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Pancreas/metabolism , Rats , Sodium-Glucose Transporter 2/physiology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
20.
Diabetes ; 69(11): 2340-2351, 2020 11.
Article in English | MEDLINE | ID: mdl-32769118

ABSTRACT

Pancreatic ß-cell proliferation has been gaining much attention as a therapeutic target for the prevention and treatment of diabetes. In order to evaluate potential ß-cell mitogens, accurate and reliable methods for the detection and quantification of the ß-cell proliferation rate are indispensable. In this study, we developed a novel tool that specifically labels replicating ß-cells as mVenus+ cells by using RIP-Cre; R26Fucci2aR mice expressing the fluorescent ubiquitination-based cell cycle indicator Fucci2a in ß-cells. In response to ß-cell proliferation stimuli, such as insulin receptor antagonist S961 and diet-induced obesity (DIO), the number of 5-ethynyl-2'-deoxyuridine-positive insulin+ cells per insulin+ cells and the number of mVenus+ cells per mCherry+ mVenus- cells + mCherry- mVenus+ cells were similarly increased in these mice. Three-dimensional imaging of optically cleared pancreas tissue from these mice enabled quantification of replicating ß-cells in the islets and morphometric analysis of the islets after known mitogenic interventions such as S961, DIO, pregnancy, and partial pancreatectomy. Thus, this novel mouse line is a powerful tool for spatiotemporal analysis and quantification of ß-cell proliferation in response to mitogenic stimulation.


Subject(s)
Cell Proliferation/physiology , Diet, High-Fat/adverse effects , Insulin-Secreting Cells/physiology , Obesity/chemically induced , Peptides/pharmacology , Animals , Cell Proliferation/drug effects , Female , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred Strains , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...