Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Diabetes ; 73(5): 806-818, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38387059

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease in which pathogenic lymphocytes target autoantigens expressed in pancreatic islets, leading to the destruction of insulin-producing ß-cells. Zinc transporter 8 (ZnT8) is a major autoantigen abundantly present on the ß-cell surface. This unique molecular target offers the potential to shield ß-cells against autoimmune attacks in T1D. Our previous work showed that a monoclonal antibody (mAb43) against cell-surface ZnT8 could home in on pancreatic islets and prevent autoantibodies from recognizing ß-cells. This study demonstrates that mAb43 binds to exocytotic sites on the ß-cell surface, masking the antigenic exposure of ZnT8 and insulin after glucose-stimulated insulin secretion. In vivo administration of mAb43 to NOD mice selectively increased the proportion of regulatory T cells in the islet, resulting in complete and sustained protection against T1D onset as well as reversal of new-onset diabetes. The mAb43-induced self-tolerance was reversible after treatment cessation, and no adverse effects were exhibited during long-term monitoring. Our findings suggest that mAb43 masking of the antigenic exposure of ß-cells suppresses the immunological cascade from B-cell antigen presentation to T cell-mediated ß-cell destruction, providing a novel islet-targeted and antigen-specific immunotherapy to prevent and reverse clinical T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Mice , Animals , Diabetes Mellitus, Type 1/metabolism , Mice, Inbred NOD , Islets of Langerhans/metabolism , Autoantigens , Insulin
2.
Front Endocrinol (Lausanne) ; 14: 1257671, 2023.
Article in English | MEDLINE | ID: mdl-37850099

ABSTRACT

Introduction: Leptin inhibits insulin secretion from isolated islets from multiple species, but the cell type that mediates this process remains elusive. Several mouse models have been used to explore this question. Ablation of the leptin receptor (Lepr) throughout the pancreatic epithelium results in altered glucose homeostasis and ex vivo insulin secretion and Ca2+ dynamics. However, Lepr removal from neither alpha nor beta cells mimics this result. Moreover, scRNAseq data has revealed an enrichment of LEPR in human islet delta cells. Methods: We confirmed LEPR upregulation in human delta cells by performing RNAseq on fixed, sorted beta and delta cells. We then used a mouse model to test whether delta cells mediate the diminished glucose-stimulated insulin secretion in response to leptin. Results: Ablation of Lepr within mouse delta cells did not change glucose homeostasis or insulin secretion, whether mice were fed a chow or high-fat diet. We further show, using a publicly available scRNAseq dataset, that islet cells expressing Lepr lie within endothelial cell clusters. Conclusions: In mice, leptin does not influence beta-cell function through delta cells.


Subject(s)
Insulin , Leptin , Animals , Humans , Mice , Glucose/metabolism , Insulin/metabolism , Leptin/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction
3.
bioRxiv ; 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36711451

ABSTRACT

The transcription factor FOXM1 regulates ß-cell proliferation and insulin secretion. Our previous work demonstrates that expressing an activated form of FOXM1 (FOXM1*) in ß cells increases ß-cell proliferation and mass in aged male mice. Additionally, FOXM1* enhances ß-cell function even in young mice, in which no ß-cell mass elevation occurs. Here, we demonstrate that FOXM1 acts in a sexually dimorphic manner in the ß cell. Expression of FOXM1* in female mouse ß cells does not affect ß-cell proliferation or glucose tolerance. Transduction of male but not female human islets with FOXM1* enhances insulin secretion in response to elevated glucose. Estrogen contributes to diabetes susceptibility differences between males and females, and the estrogen receptor (ER)α is the primary mediator of ß-cell estrogen signaling. We show that FOXM1* can rescue impaired glucose tolerance in female mice with a pancreas-wide ERα deletion. Further, FOXM1 and ERα binding sites overlap with each other and with other ß-cell-enriched transcription factors, including ISL1, PAX6, MAF, and GATA. These data indicate that FOMX1 and ERα cooperate to regulate ß-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.

4.
Diabetes ; 72(2): 184-195, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36448936

ABSTRACT

Type 1 diabetes (T1D) is a disease in which autoimmune attacks are directed at the insulin-producing ß-cell in the pancreatic islet. Autoantigens on the ß-cell surface membrane are specific markers for molecular recognition and targets for engagement by autoreactive B lymphocytes, which produce islet cell surface autoantibody (ICSA) upon activation. We report the cloning of an ICSA (mAb43) that recognizes a major T1D autoantigen, ZnT8, with a subnanomolar binding affinity and conformation specificity. We demonstrate that cell-surface binding of mAb43 protects the extracellular epitope of ZnT8 against immunolabeling by serum ICSA from a patient with T1D. Furthermore, mAb43 exhibits in vitro and ex vivo specificity for islet cells, mirroring the exquisite specificity of islet autoimmunity in T1D. Systemic administration of mAb43 yields a pancreas-specific biodistribution in mice and islet homing of an mAb43-linked imaging payload through the pancreatic vasculature, thereby validating the in vivo specificity of mAb43. Identifying ZnT8 as a major antigenic target of ICSA allows for research into the molecular recognition and engagement of autoreactive B cells in the chronic phase of T1D progression. The in vivo islet specificity of mAb43 could be further exploited to develop in vivo imaging and islet-specific immunotherapies.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Mice , Autoantibodies , Autoantigens , Diabetes Mellitus, Type 1/therapy , Tissue Distribution
5.
Cell Metab ; 34(9): 1394-1409.e4, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36070683

ABSTRACT

Three-dimensional (3D) chromatin organization maps help dissect cell-type-specific gene regulatory programs. Furthermore, 3D chromatin maps contribute to elucidating the pathogenesis of complex genetic diseases by connecting distal regulatory regions and genetic risk variants to their respective target genes. To understand the cell-type-specific regulatory architecture of diabetes risk, we generated transcriptomic and 3D epigenomic profiles of human pancreatic acinar, alpha, and beta cells using single-cell RNA-seq, single-cell ATAC-seq, and high-resolution Hi-C of sorted cells. Comparisons of these profiles revealed differential A/B (open/closed) chromatin compartmentalization, chromatin looping, and transcriptional factor-mediated control of cell-type-specific gene regulatory programs. We identified a total of 4,750 putative causal-variant-to-target-gene pairs at 194 type 2 diabetes GWAS signals using pancreatic 3D chromatin maps. We found that the connections between candidate causal variants and their putative target effector genes are cell-type stratified and emphasize previously underappreciated roles for alpha and acinar cells in diabetes pathogenesis.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Chromatin , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation , Humans , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology
6.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35642629

ABSTRACT

BACKGROUNDMultiple islet autoantibodies (AAbs) predict the development of type 1 diabetes (T1D) and hyperglycemia within 10 years. By contrast, T1D develops in only approximately 15% of individuals who are positive for single AAbs (generally against glutamic acid decarboxylase [GADA]); hence, the single GADA+ state may represent an early stage of T1D.METHODSHere, we functionally, histologically, and molecularly phenotyped human islets from nondiabetic GADA+ and T1D donors.RESULTSSimilar to the few remaining ß cells in the T1D islets, GADA+ donor islets demonstrated a preserved insulin secretory response. By contrast, α cell glucagon secretion was dysregulated in both GADA+ and T1D islets, with impaired glucose suppression of glucagon secretion. Single-cell RNA-Seq of GADA+ α cells revealed distinct abnormalities in glycolysis and oxidative phosphorylation pathways and a marked downregulation of cAMP-dependent protein kinase inhibitor ß (PKIB), providing a molecular basis for the loss of glucose suppression and the increased effect of 3-isobutyl-1-methylxanthine (IBMX) observed in GADA+ donor islets.CONCLUSIONWe found that α cell dysfunction was present during the early stages of islet autoimmunity at a time when ß cell mass was still normal, raising important questions about the role of early α cell dysfunction in the progression of T1D.FUNDINGThis work was supported by grants from the NIH (3UC4DK112217-01S1, U01DK123594-02, UC4DK112217, UC4DK112232, U01DK123716, and P30 DK019525) and the Vanderbilt Diabetes Research and Training Center (DK20593).


Subject(s)
Diabetes Mellitus, Type 1 , Glutamate Decarboxylase , Autoantibodies , Glucagon , Glucose , Humans
7.
Sleep ; 45(8)2022 08 11.
Article in English | MEDLINE | ID: mdl-35537191

ABSTRACT

We investigated the potential role of sleep-trait associated genetic loci in conferring a degree of their effect via pancreatic α- and ß-cells, given that both sleep disturbances and metabolic disorders, including type 2 diabetes and obesity, involve polygenic contributions and complex interactions. We determined genetic commonalities between sleep and metabolic disorders, conducting linkage disequilibrium genetic correlation analyses with publicly available GWAS summary statistics. Then we investigated possible enrichment of sleep-trait associated SNPs in promoter-interacting open chromatin regions within α- and ß-cells, intersecting public GWAS reports with our own ATAC-seq and high-resolution promoter-focused Capture C data generated from both sorted human α-cells and an established human beta-cell line (EndoC-ßH1). Finally, we identified putative effector genes physically interacting with sleep-trait associated variants in α- and EndoC-ßH1cells running variant-to-gene mapping and establish pathways in which these genes are significantly involved. We observed that insomnia, short and long sleep-but not morningness-were significantly correlated with type 2 diabetes, obesity and other metabolic traits. Both the EndoC-ßH1 and α-cells were enriched for insomnia loci (p = .01; p = .0076), short sleep loci (p = .017; p = .022) and morningness loci (p = 2.2 × 10-7; p = .0016), while the α-cells were also enriched for long sleep loci (p = .034). Utilizing our promoter contact data, we identified 63 putative effector genes in EndoC-ßH1 and 76 putative effector genes in α-cells, with these genes showing significant enrichment for organonitrogen and organophosphate biosynthesis, phosphatidylinositol and phosphorylation, intracellular transport and signaling, stress responses and cell differentiation. Our data suggest that a subset of sleep-related loci confer their effects via cells in pancreatic islets.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Sleep Initiation and Maintenance Disorders , Chromosome Mapping , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Islets of Langerhans/metabolism , Obesity/metabolism , Sleep , Sleep Initiation and Maintenance Disorders/metabolism
8.
Nat Metab ; 4(2): 284-299, 2022 02.
Article in English | MEDLINE | ID: mdl-35228745

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease in which immune cells destroy insulin-producing beta cells. The aetiology of this complex disease is dependent on the interplay of multiple heterogeneous cell types in the pancreatic environment. Here, we provide a single-cell atlas of pancreatic islets of 24 T1D, autoantibody-positive and nondiabetic organ donors across multiple quantitative modalities including ~80,000 cells using single-cell transcriptomics, ~7,000,000 cells using cytometry by time of flight and ~1,000,000 cells using in situ imaging mass cytometry. We develop an advanced integrative analytical strategy to assess pancreatic islets and identify canonical cell types. We show that a subset of exocrine ductal cells acquires a signature of tolerogenic dendritic cells in an apparent attempt at immune suppression in T1D donors. Our multimodal analyses delineate cell types and processes that may contribute to T1D immunopathogenesis and provide an integrative procedure for exploration and discovery of human pancreatic function.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Pancreas/metabolism , Pancreatic Hormones/metabolism
9.
Compr Physiol ; 11(3): 1961-1978, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34061978

ABSTRACT

The development and maintenance of differentiation is vital to the function of mature cells. Terminal differentiation is achieved by locking in the expression of genes essential for the function of those cells. Gene expression and its memory through generations of cell division is controlled by transcription factors and a host of epigenetic marks. In type 2 diabetes, ß cells have altered gene expression compared to controls, accompanied by altered chromatin marks. Mutations, diet, and environment can all disrupt the implementation and preservation of the distinctive ß-cell transcriptional signature. Understanding of the full complement of genomic control in ß cells is still nascent. This article describes the known effects of histone marks and variants, DNA methylation, how they are regulated in the ß cell, and how they affect cell-fate specification, maintenance, and lineage propagation. © 2021 American Physiological Society. Compr Physiol 11:1-18, 2021.


Subject(s)
Diabetes Mellitus, Type 2 , Histones , Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Humans
10.
Endocrinology ; 161(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32267917

ABSTRACT

Targeted gene ablation studies of the endocrine pancreas have long suffered from suboptimal Cre deleter strains. In many cases, Cre lines purportedly specific for beta cells also displayed expression in other islet endocrine cells or in a subset of neurons in the brain. Several pancreas and endocrine Cre lines have experienced silencing or mosaicism over time. In addition, many Cre transgenic constructs were designed to include the hGH mini-gene, which by itself increases beta-cell replication and decreases beta-cell function. More recently, driver lines with Cre or CreER inserted into the Ins1 locus were generated, with the intent of producing ß cell-specific Cre lines with faithful recapitulation of insulin expression. These lines were bred in multiple labs to several different mouse lines harboring various lox alleles. In our hands, the ability of the Ins1-Cre and Ins1-CreER lines to delete target genes varied from that originally reported, with both alleles displaying low levels of expression, increased levels of methylation compared to the wild-type allele, and ultimately inefficient or absent target deletion. Thus, caution is warranted in the interpretation of results obtained with these genetic tools, and Cre expression and activity should be monitored regularly when using these lines.


Subject(s)
DNA Methylation/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Integrases/genetics , Recombination, Genetic/genetics , Alleles , Animals , Cells, Cultured , Female , Gene Silencing , HEK293 Cells , Humans , Insulin/metabolism , Insulin-Secreting Cells/physiology , Integrases/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics
11.
Diabetes ; 67(9): 1807-1815, 2018 09.
Article in English | MEDLINE | ID: mdl-30084829

ABSTRACT

Type 2 diabetes mellitus (T2DM) is characterized by the inability of the insulin-producing ß-cells to overcome insulin resistance. We previously identified an imprinted region on chromosome 14, the DLK1-MEG3 locus, as being downregulated in islets from humans with T2DM. In this study, using targeted epigenetic modifiers, we prove that increased methylation at the promoter of Meg3 in mouse ßTC6 ß-cells results in decreased transcription of the maternal transcripts associated with this locus. As a result, the sensitivity of ß-cells to cytokine-mediated oxidative stress was increased. Additionally, we demonstrate that an evolutionarily conserved intronic region at the MEG3 locus can function as an enhancer in ßTC6 ß-cells. Using circular chromosome conformation capture followed by high-throughput sequencing, we demonstrate that the promoter of MEG3 physically interacts with this novel enhancer and other putative regulatory elements in this imprinted region in human islets. Remarkably, this enhancer is bound in an allele-specific manner by the transcription factors FOXA2, PDX1, and NKX2.2. Overall, these data suggest that the intronic MEG3 enhancer plays an important role in the regulation of allele-specific expression at the imprinted DLK1-MEG3 locus in human ß-cells, which in turn impacts the sensitivity of ß-cells to cytokine-mediated oxidative stress.


Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/metabolism , Membrane Proteins/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Animals , Calcium-Binding Proteins , Cell Line , Cytokines/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Diabetes Mellitus, Type 2/pathology , Enhancer Elements, Genetic , Epigenesis, Genetic , Genetic Loci , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/pathology , Locus Control Region , Membrane Proteins/genetics , Mice , Mutation , Nuclear Proteins , Oxidative Stress/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tissue Banks , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Mol Metab ; 6(9): 1066-1076, 2017 09.
Article in English | MEDLINE | ID: mdl-28951829

ABSTRACT

BACKGROUND: Epigenetics, in the broadest sense, governs all aspects of the life of any multicellular organism, as it controls how differentiated cells arrive at their unique phenotype during development and differentiation, despite having a uniform (with some exceptions such as T-cells and germ cells) genetic make-up. The endocrine pancreas is no exception. Transcriptional regulators and epigenetic modifiers shape the differentiation of the five major endocrine cell types from their common precursor in the fetal pancreatic bud. Beyond their role in cell differentiation, interactions of the organism with the environment are also often encoded into permanent or semi-permanent epigenetic marks and affect cellular behavior and organismal health. Epigenetics is defined as any heritable - at least through one mitotic cell division - change in phenotype or trait that is not the result of a change in genomic DNA sequence, and it forms the basis that mediates the environmental impact on diabetes susceptibility and islet function. SCOPE OF REVIEW: We will summarize the impact of epigenetic regulation on islet cell development, maturation, function, and pathophysiology. We will briefly recapitulate the major epigenetic marks and their relationship to gene activity, and outline novel strategies to employ targeted epigenetic modifications as a tool to improve islet cell function. MAJOR CONCLUSIONS: The improved understanding of the epigenetic underpinnings of islet cell differentiation, function and breakdown, as well as the development of innovative tools for their manipulation, is key to islet cell biology and the discovery of novel approaches to therapies for islet cell failure.


Subject(s)
Diabetes Mellitus/genetics , Islets of Langerhans/metabolism , Animals , Cell Differentiation/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Histones/genetics , Humans , Islets of Langerhans/physiology , Pancreas/metabolism , Phenotype , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Regulatory Elements, Transcriptional/genetics , Transcriptional Activation/genetics
13.
Diabetes Res Clin Pract ; 123: 24-36, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27918975

ABSTRACT

Type 2 diabetes is a highly heritable disease, but only ∼15% of this heritability can be explained by known genetic variant loci. In fact, body mass index is more predictive of diabetes than any of the common risk alleles identified by genome-wide association studies. This discrepancy may be explained by epigenetic inheritance, whereby changes in gene regulation can be passed along to offspring. Epigenetic changes throughout an organism's lifetime, based on environmental factors such as chemical exposures, diet, physical activity, and age, can also affect gene expression and susceptibility to diabetes. Recently, novel genome-wide assays of epigenetic marks have resulted in a greater understanding of how genetics, epigenetics, and the environment interact in the development and inheritance of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic/physiology , Insulin-Secreting Cells/metabolism , Animals , Blood Glucose/genetics , Blood Glucose/metabolism , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Humans , Inheritance Patterns , Insulin-Secreting Cells/physiology
14.
Development ; 143(24): 4558-4570, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27965437

ABSTRACT

Forkhead box (Fox) transcription factors are evolutionarily conserved in organisms ranging from yeast to humans. They regulate diverse biological processes both during development and throughout adult life. Mutations in many Fox genes are associated with human disease and, as such, various animal models have been generated to study the function of these transcription factors in mechanistic detail. In many cases, the absence of even a single Fox transcription factor is lethal. In this Primer, we provide an overview of the Fox family, highlighting several key Fox transcription factor families that are important for mammalian development.


Subject(s)
Cell Cycle/genetics , Embryonic Development/genetics , Energy Metabolism/genetics , Forkhead Transcription Factors/genetics , Animals , Humans
15.
Cell Metab ; 24(4): 616-626, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732837

ABSTRACT

The human endocrine pancreas consists of multiple cell types and plays a critical role in glucose homeostasis. Here, we apply mass cytometry technology to measure all major islet hormones, proliferative markers, and readouts of signaling pathways involved in proliferation at single-cell resolution. Using this innovative technology, we simultaneously examined baseline proliferation levels of all endocrine cell types from birth through adulthood, as well as in response to the mitogen harmine. High-dimensional analysis of our marker protein expression revealed three major clusters of beta cells within individuals. Proliferating beta cells are confined to two of the clusters.


Subject(s)
Flow Cytometry/methods , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Single-Cell Analysis/methods , Cell Aggregation/drug effects , Cell Proliferation/drug effects , Harmine/pharmacology , Humans , Islets of Langerhans/drug effects
16.
Diabetes ; 65(10): 3028-38, 2016 10.
Article in English | MEDLINE | ID: mdl-27364731

ABSTRACT

Human pancreatic islets consist of multiple endocrine cell types. To facilitate the detection of rare cellular states and uncover population heterogeneity, we performed single-cell RNA sequencing (RNA-seq) on islets from multiple deceased organ donors, including children, healthy adults, and individuals with type 1 or type 2 diabetes. We developed a robust computational biology framework for cell type annotation. Using this framework, we show that α- and ß-cells from children exhibit less well-defined gene signatures than those in adults. Remarkably, α- and ß-cells from donors with type 2 diabetes have expression profiles with features seen in children, indicating a partial dedifferentiation process. We also examined a naturally proliferating α-cell from a healthy adult, for which pathway analysis indicated activation of the cell cycle and repression of checkpoint control pathways. Importantly, this replicating α-cell exhibited activated Sonic hedgehog signaling, a pathway not previously known to contribute to human α-cell proliferation. Our study highlights the power of single-cell RNA-seq and provides a stepping stone for future explorations of cellular heterogeneity in pancreatic endocrine cells.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Transcriptome/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Computational Biology/methods , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Microfluidics/methods , Signal Transduction/genetics , Signal Transduction/physiology
17.
Cell Mol Gastroenterol Hepatol ; 2(2): 175-188, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26949732

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial stem cells that express Lgr5 and/or Bmi1 continuously replicate and generate differentiated cells throughout life1. Previously, Paneth cells were suggested to constitute an epithelium-intrinsic niche that regulates the behavior of these stem cells2. However, ablating Paneth cells has no effect on maintenance of functional stem cells3-5. Here, we demonstrate definitively that a small subset of mesenchymal, subepithelial cells expressing the winged-helix transcription factor Foxl1 are a critical component of the intestinal stem cell niche. METHODS: We genetically ablated Foxl1+ mesenchymal cells in adult mice using two separate models by expressing either the human or simian diphtheria toxin receptor (DTR) under Foxl1 promoter control. CONCLUSIONS: Killing Foxl1+ cells by diphtheria toxin administration led to an abrupt cessation of proliferation of both epithelial stem- and transit-amplifying progenitor-cell populations that was associated with a loss of active Wnt signaling to the intestinal epithelium. Therefore, Foxl1-expressing mesenchymal cells constitute the fundamental niche for intestinal stem cells.

18.
Diabetes ; 64(11): 3829-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26251404

ABSTRACT

Type 2 diabetes incidence increases with age, while ß-cell replication declines. The transcription factor FoxM1 is required for ß-cell replication in various situations, and its expression declines with age. We hypothesized that increased FoxM1 activity in aged ß-cells would rejuvenate proliferation. Induction of an activated form of FoxM1 was sufficient to increase ß-cell mass and proliferation in 12-month-old male mice after just 2 weeks. Unexpectedly, at 2 months of age, induction of activated FoxM1 in male mice improved glucose homeostasis with unchanged ß-cell mass. Cells expressing activated FoxM1 demonstrated enhanced glucose-stimulated Ca2+ influx, which resulted in improved glucose tolerance through enhanced ß-cell function. Conversely, our laboratory has previously demonstrated that mice lacking FoxM1 in the pancreas display glucose intolerance or diabetes with only a 60% reduction in ß-cell mass, suggesting that the loss of FoxM1 is detrimental to ß-cell function. Ex vivo insulin secretion was therefore examined in size-matched islets from young mice lacking FoxM1 in ß-cells. Foxm1-deficient islets indeed displayed reduced insulin secretion. Our studies reveal that activated FoxM1 increases ß-cell replication while simultaneously enhancing insulin secretion and improving glucose homeostasis, making FoxM1 an attractive therapeutic target for diabetes.


Subject(s)
Cell Proliferation/genetics , Diabetes Mellitus, Type 2/metabolism , Forkhead Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Insulin Secretion , Male , Mice
19.
Mol Endocrinol ; 28(9): 1435-47, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25073103

ABSTRACT

The forkhead box transcription factor FoxM1, a positive regulator of the cell cycle, is required for ß-cell mass expansion postnatally, during pregnancy, and after partial pancreatectomy. Up-regulation of full-length FoxM1, however, is unable to stimulate increases in ß-cell mass in unstressed mice or after partial pancreatectomy, probably due to the lack of posttranslational activation. We hypothesized that expression of an activated form of FoxM1 could aid in recovery after ß-cell injury. We therefore derived transgenic mice that inducibly express an activated version of FoxM1 in ß-cells (RIP-rtTA;TetO-hemagglutinin (HA)-Foxm1(Δ)(NRD) mice). This N-terminally truncated form of FoxM1 bypasses 2 posttranslational controls: exposure of the forkhead DNA binding domain and targeted proteasomal degradation. Transgenic mice were subjected to streptozotocin (STZ)-induced ß-cell ablation to test whether activated FoxM1 can promote ß-cell regeneration. Mice expressing HA-FoxM1(ΔNRD) displayed decreased ad libitum-fed blood glucose and increased ß-cell mass. ß-Cell proliferation was actually decreased in RIP-rtTA:TetO-HA-Foxm1(NRD) mice compared with that in RIP-rtTA mice 7 days after STZ treatment. Unexpectedly, ß-cell death was decreased 2 days after STZ treatment. RNA sequencing analysis indicated that activated FoxM1 alters the expression of extracellular matrix and immune cell gene profiles, which may protect against STZ-mediated death. These studies highlight a previously underappreciated role for FoxM1 in promoting ß-cell survival.


Subject(s)
Forkhead Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Streptozocin/chemistry , Animals , Cell Cycle , Cell Death , Cell Proliferation , Cell Survival , Diabetes Mellitus/metabolism , Female , Forkhead Box Protein M1 , Immune System , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Regeneration , Sequence Analysis, RNA
20.
Am J Physiol Endocrinol Metab ; 306(12): E1460-7, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24760991

ABSTRACT

ß-Cell mass is a parameter commonly measured in studies of islet biology and diabetes. However, the rigorous quantification of pancreatic ß-cell mass using conventional histological methods is a time-consuming process. Rapidly evolving virtual slide technology with high-resolution slide scanners and newly developed image analysis tools has the potential to transform ß-cell mass measurement. To test the effectiveness and accuracy of this new approach, we assessed pancreata from normal C57Bl/6J mice and from mouse models of ß-cell ablation (streptozotocin-treated mice) and ß-cell hyperplasia (leptin-deficient mice), using a standardized systematic sampling of pancreatic specimens. Our data indicate that automated analysis of virtual pancreatic slides is highly reliable and yields results consistent with those obtained by conventional morphometric analysis. This new methodology will allow investigators to dramatically reduce the time required for ß-cell mass measurement by automating high-resolution image capture and analysis of entire pancreatic sections.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Insulin-Secreting Cells/pathology , Models, Biological , Obesity/pathology , Pancreas/pathology , Animals , Artificial Intelligence , Automation, Laboratory , Cell Size , Computational Biology , Expert Systems , Hyperplasia , Image Processing, Computer-Assisted , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Obese , Microtomy , Pancreas/cytology , Reproducibility of Results , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...