Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Publication year range
1.
Nature ; 567(7746): 43-48, 2019 03.
Article in English | MEDLINE | ID: mdl-30760930

ABSTRACT

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Subject(s)
Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/pathology , Animals , Biomarkers/analysis , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Female , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/transplantation , Glucose/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Male , Mice , Organ Specificity/drug effects , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Proteomics , Sequence Analysis, RNA , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome , Transduction, Genetic
2.
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
3.
Stem Cells ; 38(3): 330-339, 2020 03.
Article in English | MEDLINE | ID: mdl-31722129

ABSTRACT

To date, most attention on tissue regeneration has focused on the exploration of positive cues promoting or allowing the engagement of natural cellular restoration upon injury. In contrast, the signals fostering cell identity maintenance in the vertebrate body have been poorly investigated; yet they are crucial, for their counteraction could become a powerful method to induce and modulate regeneration. Here we review the mechanisms inhibiting pro-regenerative spontaneous adaptive cell responses in different model organisms and organs. The pharmacological or genetic/epigenetic modulation of such regenerative brakes could release a dormant but innate adaptive competence of certain cell types and therefore boost tissue regeneration in different situations.


Subject(s)
Regenerative Medicine/methods , Wound Healing/physiology , Humans
4.
Nature ; 514(7523): 503-7, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25141178

ABSTRACT

Total or near-total loss of insulin-producing ß-cells occurs in type 1 diabetes. Restoration of insulin production in type 1 diabetes is thus a major medical challenge. We previously observed in mice in which ß-cells are completely ablated that the pancreas reconstitutes new insulin-producing cells in the absence of autoimmunity. The process involves the contribution of islet non-ß-cells; specifically, glucagon-producing α-cells begin producing insulin by a process of reprogramming (transdifferentiation) without proliferation. Here we show the influence of age on ß-cell reconstitution from heterologous islet cells after near-total ß-cell loss in mice. We found that senescence does not alter α-cell plasticity: α-cells can reprogram to produce insulin from puberty through to adulthood, and also in aged individuals, even a long time after ß-cell loss. In contrast, before puberty there is no detectable α-cell conversion, although ß-cell reconstitution after injury is more efficient, always leading to diabetes recovery. This process occurs through a newly discovered mechanism: the spontaneous en masse reprogramming of somatostatin-producing δ-cells. The juveniles display 'somatostatin-to-insulin' δ-cell conversion, involving dedifferentiation, proliferation and re-expression of islet developmental regulators. This juvenile adaptability relies, at least in part, upon the combined action of FoxO1 and downstream effectors. Restoration of insulin producing-cells from non-ß-cell origins is thus enabled throughout life via δ- or α-cell spontaneous reprogramming. A landscape with multiple intra-islet cell interconversion events is emerging, offering new perspectives for therapy.


Subject(s)
Aging/physiology , Cell Transdifferentiation , Diabetes Mellitus, Experimental/pathology , Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Regeneration , Somatostatin-Secreting Cells/cytology , Animals , Cell Dedifferentiation , Cell Proliferation , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Sexual Maturation , Somatostatin/biosynthesis , Somatostatin/metabolism , Somatostatin-Secreting Cells/metabolism
5.
Genes Dev ; 25(16): 1680-5, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21852533

ABSTRACT

Using single transcription factors to reprogram cells could produce important insights into the epigenetic mechanisms that direct normal differentiation, or counter inappropriate plasticity, or even provide new ways of manipulating normal ontogeny in vitro to control lineage diversification and differentiation. We enforced Pdx1 expression from the Neurogenin-3-expressing endocrine commitment point onward and found during the embryonic period a minor increased ß-cell allocation with accompanying reduced α-cell numbers. More surprisingly, almost all remaining Pdx1-containing glucagon/Arx-producing cells underwent a fairly rapid conversion at postnatal stages, through glucagon-insulin double positivity, to a state indistinguishable from normal ß cells, resulting in complete α-cell absence. This α-to-ß conversion was not caused by activating Pdx1 in the later glucagon-expressing state. Our findings reveal that Pdx1 can work single-handedly as a potent context-dependent autonomous reprogramming agent, and suggest a postnatal differentiation evaluation stage involved in normal endocrine maturation.


Subject(s)
Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glucagon/genetics , Glucagon/metabolism , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/genetics , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/embryology , Pancreas/growth & development , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
6.
Diabetologia ; 60(6): 1043-1050, 2017 06.
Article in English | MEDLINE | ID: mdl-28343277

ABSTRACT

AIMS/HYPOTHESIS: Transcription factor 7-like 2 (TCF7L2) is a high mobility group (HMG) box-containing transcription factor and downstream effector of the Wnt signalling pathway. SNPs in the TCF7L2 gene have previously been associated with an increased risk of type 2 diabetes in genome-wide association studies. In animal studies, loss of Tcf7l2 function is associated with defective islet beta cell function and survival. Here, we explore the role of TCF7L2 in the control of the counter-regulatory response to hypoglycaemia by generating mice with selective deletion of the Tcf7l2 gene in pancreatic alpha cells. METHODS: Alpha cell-selective deletion of Tcf7l2 was achieved by crossing mice with floxed Tcf7l2 alleles to mice bearing a Cre recombinase transgene driven by the preproglucagon promoter (PPGCre), resulting in Tcf7l2AKO mice. Glucose homeostasis and hormone secretion in vivo and in vitro, and islet cell mass were measured using standard techniques. RESULTS: While glucose tolerance was unaffected in Tcf7l2AKO mice, glucose infusion rates were increased (AUC for glucose during the first 60 min period of hyperinsulinaemic-hypoglycaemic clamp test was increased by 1.98 ± 0.26-fold [p < 0.05; n = 6] in Tcf7l2AKO mice vs wild-type mice) and glucagon secretion tended to be lower (plasma glucagon: 0.40 ± 0.03-fold vs wild-type littermate controls [p < 0.01; n = 6]). Tcf7l2AKO mice displayed reduced fasted plasma glucose concentration. Glucagon release at low glucose was impaired in islets isolated from Tcf7l2AKO mice (0.37 ± 0.02-fold vs islets from wild-type littermate control mice [p < 0.01; n = 6). Alpha cell mass was also reduced (72.3 ± 20.3% [p < 0.05; n = 7) in Tcf7l2AKO mice compared with wild-type mice. CONCLUSIONS/INTERPRETATION: The present findings demonstrate an alpha cell-autonomous role for Tcf7l2 in the control of pancreatic glucagon secretion and the maintenance of alpha cell mass and function.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Hypoglycemia/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Female , Glucose/metabolism , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Knockout , Proglucagon/genetics , Real-Time Polymerase Chain Reaction , Transcription Factor 7-Like 2 Protein/genetics
7.
Sensors (Basel) ; 17(3)2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28335445

ABSTRACT

This work presents a novel strategy to decipher fragments of Egyptian cartouches identifying the hieroglyphs of which they are composed. A cartouche is a drawing, usually inside an oval, that encloses a group of hieroglyphs representing the name of a monarch. Aiming to identify these drawings, the proposed method is based on several techniques frequently used in computer vision and consists of three main stages: first, a picture of the cartouche is taken as input and its contour is localized. In the second stage, each hieroglyph is individually extracted and identified. Finally, the cartouche is interpreted: the sequence of the hieroglyphs is established according to a previously generated benchmark. This sequence corresponds to the name of the king. Although this method was initially conceived to deal with both high and low relief writing in stone, it can be also applied to painted hieroglyphs. This approach is not affected by variable lighting conditions, or the intensity and the completeness of the objects. This proposal has been tested on images obtained from the Abydos King List and other Egyptian monuments and archaeological excavations. The promising results give new possibilities to recognize hieroglyphs, opening a new way to decipher longer texts and inscriptions, being particularly useful in museums and Egyptian environments. Additionally, devices used for acquiring visual information from cartouches (i.e., smartphones), can be part of a navigation system for museums where users are located in indoor environments by means of the combination of WiFi Positioning Systems (WPS) and depth cameras, as unveiled at the end of the document.

8.
Diabetologia ; 59(9): 1938-47, 2016 09.
Article in English | MEDLINE | ID: mdl-27338626

ABSTRACT

AIMS/HYPOTHESIS: Per-Arnt-Sim kinase (PASK) is a nutrient-regulated domain-containing protein kinase previously implicated in the control of insulin gene expression and glucagon secretion. Here, we explore the roles of PASK in the control of islet hormone release, by generating mice with selective deletion of the Pask gene in pancreatic beta or alpha cells. METHODS: Floxed alleles of Pask were produced by homologous recombination and animals bred with mice bearing beta (Ins1 (Cre); PaskBKO) or alpha (Ppg (Cre) [also known as Gcg]; PaskAKO) cell-selective Cre recombinase alleles. Glucose homeostasis and hormone secretion in vivo and in vitro, gene expression and islet cell mass were measured using standard techniques. RESULTS: Ins1 (Cre)-based recombination led to efficient beta cell-targeted deletion of Pask. Beta cell mass was reduced by 36.5% (p < 0.05) compared with controls in PaskBKO mice, as well as in global Pask-null mice (38%, p < 0.05). PaskBKO mice displayed normal body weight and fasting glycaemia, but slightly impaired glucose tolerance, and beta cell proliferation, after maintenance on a high-fat diet. Whilst glucose tolerance was unaffected in PaskAKO mice, glucose infusion rates were increased, and glucagon secretion tended to be lower, during hypoglycaemic clamps. Although alpha cell mass was increased (21.9%, p < 0.05), glucagon release at low glucose was impaired (p < 0.05) in PaskAKO islets. CONCLUSIONS/INTERPRETATION: The findings demonstrate cell-autonomous roles for PASK in the control of pancreatic endocrine hormone secretion. Differences between the glycaemic phenotype of global vs cell type-specific null mice suggest important roles for tissue interactions in the control of glycaemia by PASK.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Diet, High-Fat/adverse effects , Glucose/metabolism , Homeostasis/genetics , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
9.
J Biol Chem ; 290(35): 21432-42, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26178371

ABSTRACT

SLC30A8 encodes a zinc transporter ZnT8 largely restricted to pancreatic islet ß- and α-cells, and responsible for zinc accumulation into secretory granules. Although common SLC30A8 variants, believed to reduce ZnT8 activity, increase type 2 diabetes risk in humans, rare inactivating mutations are protective. To investigate the role of Slc30a8 in the control of glucagon secretion, Slc30a8 was inactivated selectively in α-cells by crossing mice with alleles floxed at exon 1 to animals expressing Cre recombinase under the pre-proglucagon promoter. Further crossing to Rosa26:tdRFP mice, and sorting of RFP(+): glucagon(+) cells from KO mice, revealed recombination in ∼ 30% of α-cells, of which ∼ 50% were ZnT8-negative (14 ± 1.8% of all α-cells). Although glucose and insulin tolerance were normal, female αZnT8KO mice required lower glucose infusion rates during hypoglycemic clamps and displayed enhanced glucagon release (p < 0.001) versus WT mice. Correspondingly, islets isolated from αZnT8KO mice secreted more glucagon at 1 mm glucose, but not 17 mm glucose, than WT controls (n = 5; p = 0.008). Although the expression of other ZnT family members was unchanged, cytoplasmic (n = 4 mice per genotype; p < 0.0001) and granular (n = 3, p < 0.01) free Zn(2+) levels were significantly lower in KO α-cells versus control cells. In response to low glucose, the amplitude and frequency of intracellular Ca(2+) increases were unchanged in α-cells of αZnT8KO KO mice. ZnT8 is thus important in a subset of α-cells for normal responses to hypoglycemia and acts via Ca(2+)-independent mechanisms.


Subject(s)
Cation Transport Proteins/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Hypoglycemia/metabolism , Animals , Cation Transport Proteins/analysis , Cation Transport Proteins/genetics , Cells, Cultured , Female , Gene Deletion , Glucagon-Secreting Cells/cytology , Glucose/metabolism , Hypoglycemia/genetics , Insulin Resistance , Mice, Inbred C57BL , Zinc/metabolism , Zinc Transporter 8
10.
Nature ; 464(7292): 1149-54, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20364121

ABSTRACT

Pancreatic insulin-producing beta-cells have a long lifespan, such that in healthy conditions they replicate little during a lifetime. Nevertheless, they show increased self-duplication after increased metabolic demand or after injury (that is, beta-cell loss). It is not known whether adult mammals can differentiate (regenerate) new beta-cells after extreme, total beta-cell loss, as in diabetes. This would indicate differentiation from precursors or another heterologous (non-beta-cell) source. Here we show beta-cell regeneration in a transgenic model of diphtheria-toxin-induced acute selective near-total beta-cell ablation. If given insulin, the mice survived and showed beta-cell mass augmentation with time. Lineage-tracing to label the glucagon-producing alpha-cells before beta-cell ablation tracked large fractions of regenerated beta-cells as deriving from alpha-cells, revealing a previously disregarded degree of pancreatic cell plasticity. Such inter-endocrine spontaneous adult cell conversion could be harnessed towards methods of producing beta-cells for diabetes therapies, either in differentiation settings in vitro or in induced regeneration.


Subject(s)
Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , Animals , Biomarkers/metabolism , Cell Count , Cell Death/drug effects , Cell Lineage , Cell Proliferation , Cellular Reprogramming , Diphtheria Toxin/pharmacology , Diphtheria Toxin/toxicity , Female , Glucagon/biosynthesis , Glucagon/genetics , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Rats , Regeneration/physiology
11.
PLoS Genet ; 9(1): e1003160, 2013.
Article in English | MEDLINE | ID: mdl-23300479

ABSTRACT

Mouse sex determination provides an attractive model to study how regulatory genetic networks and signaling pathways control cell specification and cell fate decisions. This study characterizes in detail the essential role played by the insulin receptor (INSR) and the IGF type I receptor (IGF1R) in adrenogenital development and primary sex determination. Constitutive ablation of insulin/IGF signaling pathway led to reduced proliferation rate of somatic progenitor cells in both XX and XY gonads prior to sex determination together with the downregulation of hundreds of genes associated with the adrenal, testicular, and ovarian genetic programs. These findings indicate that prior to sex determination somatic progenitors in Insr;Igf1r mutant gonads are not lineage primed and thus incapable of upregulating/repressing the male and female genetic programs required for cell fate restriction. In consequence, embryos lacking functional insulin/IGF signaling exhibit (i) complete agenesis of the adrenal cortex, (ii) embryonic XY gonadal sex reversal, with a delay of Sry upregulation and the subsequent failure of the testicular genetic program, and (iii) a delay in ovarian differentiation so that Insr;Igf1r mutant gonads, irrespective of genetic sex, remained in an extended undifferentiated state, before the ovarian differentiation program ultimately is initiated at around E16.5.


Subject(s)
Gonads , Insulin , Receptor, IGF Type 1 , Receptor, Insulin , Sex Determination Processes/genetics , Adrenal Cortex/growth & development , Adrenal Cortex/pathology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , Disorders of Sex Development/genetics , Female , Gonads/growth & development , Gonads/metabolism , Gonads/pathology , Humans , Insulin/genetics , Insulin/metabolism , Male , Mice , Ovary/growth & development , Ovary/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sex Chromosomes , Signal Transduction , Testis/growth & development , Testis/metabolism
12.
PLoS Genet ; 9(1): e1003274, 2013.
Article in English | MEDLINE | ID: mdl-23382704

ABSTRACT

All pancreatic endocrine cell types arise from a common endocrine precursor cell population, yet the molecular mechanisms that establish and maintain the unique gene expression programs of each endocrine cell lineage have remained largely elusive. Such knowledge would improve our ability to correctly program or reprogram cells to adopt specific endocrine fates. Here, we show that the transcription factor Nkx6.1 is both necessary and sufficient to specify insulin-producing beta cells. Heritable expression of Nkx6.1 in endocrine precursors of mice is sufficient to respecify non-beta endocrine precursors towards the beta cell lineage, while endocrine precursor- or beta cell-specific inactivation of Nkx6.1 converts beta cells to alternative endocrine lineages. Remaining insulin(+) cells in conditional Nkx6.1 mutants fail to express the beta cell transcription factors Pdx1 and MafA and ectopically express genes found in non-beta endocrine cells. By showing that Nkx6.1 binds to and represses the alpha cell determinant Arx, we identify Arx as a direct target of Nkx6.1. Moreover, we demonstrate that Nkx6.1 and the Arx activator Isl1 regulate Arx transcription antagonistically, thus establishing competition between Isl1 and Nkx6.1 as a critical mechanism for determining alpha versus beta cell identity. Our findings establish Nkx6.1 as a beta cell programming factor and demonstrate that repression of alternative lineage programs is a fundamental principle by which beta cells are specified and maintained. Given the lack of Nkx6.1 expression and aberrant activation of non-beta endocrine hormones in human embryonic stem cell (hESC)-derived insulin(+) cells, our study has significant implications for developing cell replacement therapies.


Subject(s)
Endocrine Cells , Homeodomain Proteins , Insulin-Secreting Cells , Insulin , Animals , Cell Differentiation/genetics , Cell Lineage , Cell- and Tissue-Based Therapy , Endocrine Cells/cytology , Endocrine Cells/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Pancreas/cytology , Stem Cells , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Diabetologia ; 58(2): 304-12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25413047

ABSTRACT

AIMS/HYPOTHESIS: Non-invasive imaging of beta cells is a much-needed development but is one that faces significant biological and technological hurdles. A relevant imaging method should at least allow for an evaluation over time of the mass of beta cells under physiological and pathological conditions, and for an assessment of novel therapies. We, therefore, investigated the ability of a new MRI probe to repeatedly measure the loss of beta cells in a rodent model. METHODS: We developed an innovative nanoparticle probe that targets the glucagon-like peptide 1 receptor, and can be used for both fluorescence imaging and MRI. Using fluorescence, we characterised the specificity and biodistribution of the probe. Using 1.5 T MRI, we longitudinally imaged the changes in insulin content in male and female mice of the RIP-DTr strain, which mimic the changes expected in type 1 and type 2 diabetes, respectively. RESULTS: We showed that this probe selectively labelled beta cells in situ, imaged in vivo native pancreatic islets and evaluated their loss after diphtheria toxin administration, in a model of graded beta cell deletion. Thus, using clinical MRI, the probe quantitatively differentiates, in the same mouse strain, between female animals featuring a 50% loss of beta cells and the males featuring an almost complete loss of beta cells. CONCLUSIONS/INTERPRETATION: The approach addresses several of the hurdles that have so far limited the non-invasive imaging of beta cells, including the potential to repeatedly monitor the very same animals using clinically available equipment, and to differentiate graded losses of beta cells.


Subject(s)
Glucagon/metabolism , Insulin-Secreting Cells/metabolism , Magnetic Resonance Imaging , Peptide Fragments/metabolism , Receptors, Glucagon/metabolism , Animals , Female , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Molecular Probes , Tissue Distribution
14.
Nat Genet ; 38(5): 589-93, 2006 May.
Article in English | MEDLINE | ID: mdl-16642023

ABSTRACT

The total mass of islets of Langerhans is reduced in individuals with type 2 diabetes, possibly contributing to the pathogenesis of this condition. Although the regulation of islet mass is complex, recent studies have suggested the importance of a signaling pathway that includes the insulin or insulin-like growth factor-1 receptors, insulin receptor substrate and phosphatidylinositol (PI) 3-kinase. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) is a serine-threonine kinase that mediates signaling downstream of PI 3-kinase. Here we show that mice that lack PDK1 specifically in pancreatic beta cells (betaPdk1-/- mice) develop progressive hyperglycemia as a result of a loss of islet mass. The mice show reductions in islet density as well as in the number and size of cells. Haploinsufficiency of the gene for the transcription factor Foxo1 resulted in a marked increase in the number, but not the size, of cells and resulted in the restoration of glucose homeostasis in betaPdk1-/- mice. These results suggest that PDK1 is important in maintenance of pancreatic cell mass and glucose homeostasis.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Protein Serine-Threonine Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Mice , Mice, Knockout , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 108(51): 20719-24, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143775

ABSTRACT

We combined multimodal imaging (bioluminescence, X-ray computed tomography, and PET), tomographic reconstruction of bioluminescent sources, and two unique, complementary models to evaluate three previously synthesized PET radiotracers thought to target pancreatic beta cells. The three radiotracers {[(18)F]fluoropropyl-(+)-dihydrotetrabenazine ([(18)F]FP-DTBZ), [(18)F](+)-2-oxiranyl-3-isobutyl-9-(3-fluoropropoxy)-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinoline ((18)F-AV-266), and (2S,3R,11bR)-9-(3-fluoropropoxy)-2-(hydroxymethyl)-3-isobutyl-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-2-ol ((18)F-AV-300)} bind vesicular monoamine transporter 2. Tomographic reconstruction of the bioluminescent signal in mice expressing luciferase only in pancreatic beta cells was used to delineate the pancreas and was coregistered with PET and X-ray computed tomography images. This strategy enabled unambiguous identification of the pancreas on PET images, permitting accurate quantification of the pancreatic PET signal. We show here that, after conditional, specific, and rapid mouse beta-cell ablation, beta-cell loss was detected by bioluminescence imaging but not by PET imaging, given that the pancreatic signal provided by three PET radiotracers was not altered. To determine whether these ligands bound human beta cells in vivo, we imaged mice transplanted with luciferase-expressing human islets. The human islets were imaged by bioluminescence but not with the PET ligands, indicating that these vesicular monoamine transporter 2-directed ligands did not specifically bind beta cells. These data demonstrate the utility of coregistered multimodal imaging as a platform for evaluation and validation of candidate ligands for imaging islets.


Subject(s)
Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Animals , Diabetes Mellitus/metabolism , Diagnostic Imaging/methods , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Ligands , Luminescence , Male , Mice , Mice, Inbred NOD , Rats , Tissue Distribution
16.
Sensors (Basel) ; 14(8): 15304-24, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25195854

ABSTRACT

An important objective in weed management is the discrimination between grasses (monocots) and broad-leaved weeds (dicots), because these two weed groups can be appropriately controlled by specific herbicides. In fact, efficiency is higher if selective treatment is performed for each type of infestation instead of using a broadcast herbicide on the whole surface. This work proposes a strategy where weeds are characterised by a set of shape descriptors (the seven Hu moments and six geometric shape descriptors). Weeds appear in outdoor field images which display real situations obtained from a RGB camera. Thus, images present a mixture of both weed species under varying conditions of lighting. In the presented approach, four decision-making methods were adapted to use the best shape descriptors as attributes and a choice was taken. This proposal establishes a novel methodology with a high success rate in weed species discrimination.


Subject(s)
Fuzzy Logic , Weed Control , Zea mays/physiology , Agriculture , Decision Making , Plant Weeds/physiology
17.
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.

18.
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
19.
Nat Metab ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169271

ABSTRACT

Insulin-producing ß-cells in pancreatic islets are regulated by systemic cues and, locally, by adjacent islet hormone-producing 'non-ß-cells' (namely α-cells, δ-cells and γ-cells). Yet whether the non-ß-cells are required for accurate insulin secretion is unclear. Here, we studied mice in which adult islets are exclusively composed of ß-cells and human pseudoislets containing only primary ß-cells. Mice lacking non-ß-cells had optimal blood glucose regulation, enhanced glucose tolerance, insulin sensitivity and restricted body weight gain under a high-fat diet. The insulin secretion dynamics in islets composed of only ß-cells was comparable to that in intact islets. Similarly, human ß-cell pseudoislets retained the glucose-regulated mitochondrial respiration, insulin secretion and exendin-4 responses of entire islets. The findings indicate that non-ß-cells are dispensable for blood glucose homeostasis and ß-cell function. These results support efforts aimed at developing diabetes treatments by generating ß-like clusters devoid of non-ß-cells, such as from pluripotent stem cells differentiated in vitro or by reprograming non-ß-cells into insulin producers in situ.

20.
Commun Biol ; 7(1): 833, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982170

ABSTRACT

Regeneration, the ability to replace injured tissues and organs, is a phenomenon commonly associated with lower vertebrates but is also observed in mammals, in specific tissues. In this study, we investigated the regenerative potential of pancreatic islets following moderate beta-cell loss in mice. Using a rapid model of moderate ablation, we observed a compensatory response characterized by transient inflammation and proliferation signatures, ultimately leading to the recovery of beta-cell identity and function. Interestingly, this proliferative response occurred independently of inflammation, as demonstrated in ablated immunodeficient mice. Furthermore, exposure to high-fat diet stimulated beta-cell proliferation but negatively impacted beta-cell function. In contrast, an equivalent slower ablation model revealed a delayed but similar proliferative response, suggesting proliferation as a common regenerative response. However, high-fat diet failed to promote proliferation in this model, indicating a differential response to metabolic stressors. Overall, our findings shed light on the complex interplay between beta-cell loss, inflammation, and stress in modulating pancreatic islet regeneration. Understanding these mechanisms could pave the way for novel therapeutic strategies based on beta-cell proliferation.


Subject(s)
Cell Proliferation , Diet, High-Fat , Insulin-Secreting Cells , Regeneration , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Mice , Diet, High-Fat/adverse effects , Male , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/pathology
SELECTION OF CITATIONS
SEARCH DETAIL