Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
Add more filters

Publication year range
1.
Cell ; 156(1-2): 304-16, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439384

ABSTRACT

A clear relationship exists between visceral obesity and type 2 diabetes, whereas subcutaneous obesity is comparatively benign. Here, we show that adipocyte-specific deletion of the coregulatory protein PRDM16 caused minimal effects on classical brown fat but markedly inhibited beige adipocyte function in subcutaneous fat following cold exposure or ß3-agonist treatment. These animals developed obesity on a high-fat diet, with severe insulin resistance and hepatic steatosis. They also showed altered fat distribution with markedly increased subcutaneous adiposity. Subcutaneous adipose tissue in mutant mice acquired many key properties of visceral fat, including decreased thermogenic and increased inflammatory gene expression and increased macrophage accumulation. Transplantation of subcutaneous fat into mice with diet-induced obesity showed a loss of metabolic benefit when tissues were derived from PRDM16 mutant animals. These findings indicate that PRDM16 and beige adipocytes are required for the "browning" of white fat and the healthful effects of subcutaneous adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , DNA-Binding Proteins/metabolism , Obesity/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Animals , DNA-Binding Proteins/genetics , Diet, High-Fat , Insulin Resistance , Mice , Mice, Knockout , Transcription Factors/genetics
2.
Mol Cell ; 74(6): 1123-1137.e6, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31053472

ABSTRACT

Abnormal processing of stressed replication forks by nucleases can cause fork collapse, genomic instability, and cell death. Despite its importance, it is poorly understood how the cell properly controls nucleases to prevent detrimental fork processing. Here, we report a signaling pathway that controls the activity of exonuclease Exo1 to prevent aberrant fork resection during replication stress. Our results indicate that replication stress elevates intracellular Ca2+ concentration ([Ca2+]i), leading to activation of CaMKK2 and the downstream kinase 5' AMP-activated protein kinase (AMPK). Following activation, AMPK directly phosphorylates Exo1 at serine 746 to promote 14-3-3 binding and inhibit Exo1 recruitment to stressed replication forks, thereby avoiding unscheduled fork resection. Disruption of this signaling pathway results in excessive ssDNA, chromosomal instability, and hypersensitivity to replication stress inducers. These findings reveal a link between [Ca2+]i and the replication stress response as well as a function of the Ca2+-CaMKK2-AMPK signaling axis in safeguarding fork structure to maintain genome stability.


Subject(s)
AMP-Activated Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium/metabolism , DNA Repair Enzymes/genetics , DNA Repair , DNA Replication , Exodeoxyribonucleases/genetics , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Calcium Signaling/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Chromatin/chemistry , Chromatin/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 120(22): e2302624120, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37205712

ABSTRACT

Human islet primary cilia are vital glucose-regulating organelles whose structure remains uncharacterized. Scanning electron microscopy (SEM) is a useful technique for studying the surface morphology of membrane projections like cilia, but conventional sample preparation does not reveal the submembrane axonemal structure, which holds key implications for ciliary function. To overcome this challenge, we combined SEM with membrane-extraction techniques to examine primary cilia in native human islets. Our data show well-preserved cilia subdomains which demonstrate both expected and unexpected ultrastructural motifs. Morphometric features were quantified when possible, including axonemal length and diameter, microtubule conformations, and chirality. We further describe a ciliary ring, a structure that may be a specialization in human islets. Key findings are correlated with fluorescence microscopy and interpreted in the context of cilia function as a cellular sensor and communications locus in pancreatic islets.


Subject(s)
Cilia , Islets of Langerhans , Humans , Microscopy, Electron, Scanning , Cilia/physiology , Microscopy, Fluorescence , Microtubules
4.
J Biol Chem ; 299(11): 105334, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37827287

ABSTRACT

Just under the plasma membrane of most animal cells lies a dense meshwork of actin filaments called the cortical cytoskeleton. In insulin-secreting pancreatic ß cells, a long-standing model posits that the cortical actin layer primarily acts to restrict access of insulin granules to the plasma membrane. Here we test this model and find that stimulating ß cells with pro-secretory stimuli (glucose and/or KCl) has little impact on the cortical actin layer. Chemical perturbations of actin polymerization, by either disrupting or enhancing filamentation, dramatically enhance glucose-stimulated insulin secretion. Using scanning electron microscopy, we directly visualize the cortical cytoskeleton, allowing us to validate the effect of these filament-disrupting chemicals. We find the state of the cortical actin layer does not correlate with levels of insulin secretion, suggesting filament disruptors act on insulin secretion independently of the cortical cytoskeleton.


Subject(s)
Actin Cytoskeleton , Actins , Insulin Secretion , Insulin-Secreting Cells , Animals , Actin Cytoskeleton/metabolism , Actins/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/metabolism
5.
Genes Dev ; 30(16): 1852-65, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27585590

ABSTRACT

The current model for endocrine cell specification in the pancreas invokes high-level production of the transcription factor Neurogenin 3 (Neurog3) in Sox9(+) bipotent epithelial cells as the trigger for endocrine commitment, cell cycle exit, and rapid delamination toward proto-islet clusters. This model posits a transient Neurog3 expression state and short epithelial residence period. We show, however, that a Neurog3(TA.LO) cell population, defined as Neurog3 transcriptionally active and Sox9(+) and often containing nonimmunodetectable Neurog3 protein, has a relatively high mitotic index and prolonged epithelial residency. We propose that this endocrine-biased mitotic progenitor state is functionally separated from a pro-ductal pool and endows them with long-term capacity to make endocrine fate-directed progeny. A novel BAC transgenic Neurog3 reporter detected two types of mitotic behavior in Sox9(+) Neurog3(TA.LO) progenitors, associated with progenitor pool maintenance or derivation of endocrine-committed Neurog3(HI) cells, respectively. Moreover, limiting Neurog3 expression dramatically increased the proportional representation of Sox9(+) Neurog3(TA.LO) progenitors, with a doubling of its mitotic index relative to normal Neurog3 expression, suggesting that low Neurog3 expression is a defining feature of this cycling endocrine-biased state. We propose that Sox9(+) Neurog3(TA.LO) endocrine-biased progenitors feed production of Neurog3(HI) endocrine-committed cells during pancreas organogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endocrine Cells/cytology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation/genetics , Mice , Mitosis , Pancreas/cytology
6.
Proc Natl Acad Sci U S A ; 117(16): 8912-8923, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253320

ABSTRACT

Pancreatic islets regulate glucose homeostasis through coordinated actions of hormone-secreting cells. What underlies the function of the islet as a unit is the close approximation and communication among heterogeneous cell populations, but the structural mediators of islet cellular cross talk remain incompletely characterized. We generated mice specifically lacking ß-cell primary cilia, a cellular organelle that has been implicated in regulating insulin secretion, and found that the ß-cell cilia are required for glucose sensing, calcium influx, insulin secretion, and cross regulation of α- and δ-cells. Protein expression profiling in islets confirms perturbation in these cellular processes and reveals additional targets of cilia-dependent signaling. At the organism level, the deletion of ß-cell cilia disrupts circulating hormone levels, impairs glucose homeostasis and fuel usage, and leads to the development of diabetes. Together, these findings demonstrate that primary cilia not only orchestrate ß-cell-intrinsic activity but also mediate cross talk both within the islet and from islets to other metabolic tissues, thus providing a unique role of cilia in nutrient metabolism and insight into the pathophysiology of diabetes.


Subject(s)
Cilia/metabolism , Diabetes Mellitus/pathology , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Calcium/metabolism , Cell Communication/physiology , Cilia/genetics , Cilia/pathology , Diabetes Mellitus/genetics , Disease Models, Animal , Energy Metabolism/physiology , Female , Glucagon-Secreting Cells/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 116(47): 23822-23828, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31694884

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a critical coenzyme for cellular energy metabolism. The aim of the present study was to determine the importance of brown and white adipose tissue (BAT and WAT) NAD+ metabolism in regulating whole-body thermogenesis and energy metabolism. Accordingly, we generated and analyzed adipocyte-specific nicotinamide phosphoribosyltransferase (Nampt) knockout (ANKO) and brown adipocyte-specific Nampt knockout (BANKO) mice because NAMPT is the rate-limiting NAD+ biosynthetic enzyme. We found ANKO mice, which lack NAMPT in both BAT and WAT, had impaired gene programs involved in thermogenesis and mitochondrial function in BAT and a blunted thermogenic (rectal temperature, BAT temperature, and whole-body oxygen consumption) response to acute cold exposure, prolonged fasting, and administration of ß-adrenergic agonists (norepinephrine and CL-316243). In addition, the absence of NAMPT in WAT markedly reduced adrenergic-mediated lipolytic activity, likely through inactivation of the NAD+-SIRT1-caveolin-1 axis, which limits an important fuel source fatty acid for BAT thermogenesis. These metabolic abnormalities were rescued by treatment with nicotinamide mononucleotide (NMN), which bypasses the block in NAD+ synthesis induced by NAMPT deficiency. Although BANKO mice, which lack NAMPT in BAT only, had BAT cellular alterations similar to the ANKO mice, BANKO mice had normal thermogenic and lipolytic responses. We also found NAMPT expression in supraclavicular adipose tissue (where human BAT is localized) obtained from human subjects increased during cold exposure, suggesting our finding in rodents could apply to people. These results demonstrate that adipose NAMPT-mediated NAD+ biosynthesis is essential for regulating adaptive thermogenesis, lipolysis, and whole-body energy metabolism.


Subject(s)
Adaptation, Physiological , Adipose Tissue, Brown/metabolism , Energy Metabolism , Homeostasis , NAD/biosynthesis , Thermogenesis , Adipose Tissue, Brown/enzymology , Animals , Caveolin 1/antagonists & inhibitors , Cold Temperature , Cytokines/genetics , Fasting , Humans , Mice , Mice, Knockout , Nicotinamide Mononucleotide/administration & dosage , Nicotinamide Phosphoribosyltransferase/genetics
8.
Proc Natl Acad Sci U S A ; 115(6): E1221-E1228, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29358401

ABSTRACT

Motile cilia are characterized by dynein motor units, which preassemble in the cytoplasm before trafficking into the cilia. Proteins required for dynein preassembly were discovered by finding human mutations that result in absent ciliary motors, but little is known about their expression, function, or interactions. By monitoring ciliogenesis in primary airway epithelial cells and MCIDAS-regulated induced pluripotent stem cells, we uncovered two phases of expression of preassembly proteins. An early phase, composed of HEATR2, SPAG1, and DNAAF2, preceded other preassembly proteins and was independent of MCIDAS regulation. The early preassembly proteins colocalized within perinuclear foci that also contained dynein arm proteins. These proteins also interacted based on immunoprecipitation and Förster resonance energy transfer (FRET) studies. FRET analysis of HEAT domain deletions and human mutations showed that HEATR2 interacted with itself and SPAG1 at multiple HEAT domains, while DNAAF2 interacted with SPAG1. Human mutations in HEATR2 did not affect this interaction, but triggered the formation of p62/Sequestosome-1-positive aggregates containing the early preassembly proteins, suggesting that degradation of an early preassembly complex is responsible for disease and pointing to key regions required for HEATR2 scaffold stability. We speculate that HEATR2 is an early scaffold for the initiation of dynein complex assembly in motile cilia.


Subject(s)
Antigens, Surface/metabolism , Cilia/physiology , GTP-Binding Proteins/metabolism , Induced Pluripotent Stem Cells/physiology , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , Respiratory Mucosa/physiology , Animals , Antigens, Surface/genetics , Axonemal Dyneins , Cells, Cultured , GTP-Binding Proteins/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Microtubule-Associated Proteins/genetics , Mutation , Phenotype , Proteins/genetics , Respiratory Mucosa/cytology
9.
Lab Invest ; 100(6): 849-862, 2020 06.
Article in English | MEDLINE | ID: mdl-32060407

ABSTRACT

Wolfram Syndrome 1 (WFS1) protein is an endoplasmic reticulum (ER) factor whose deficiency results in juvenile-onset diabetes secondary to cellular dysfunction and apoptosis. The mechanisms guiding ß-cell outcomes secondary to WFS1 function, however, remain unclear. Here, we show that WFS1 preserves normal ß-cell physiology by promoting insulin biosynthesis and negatively regulating ER stress. Depletion of Wfs1 in vivo and in vitro causes functional defects in glucose-stimulated insulin secretion and insulin content, triggering Chop-mediated apoptotic pathways. Genetic proof of concept studies coupled with RNA-seq reveal that increasing WFS1 confers a functional and a survival advantage to ß-cells under ER stress by increasing insulin gene expression and downregulating the Chop-Trib3 axis, thereby activating Akt pathways. Remarkably, WFS1 and INS levels are reduced in type-2 diabetic (T2DM) islets, suggesting that WFS1 may contribute to T2DM ß-cell pathology. Taken together, this work reveals essential pathways regulated by WFS1 to control ß-cell survival and function primarily through preservation of ER homeostasis.


Subject(s)
Insulin-Secreting Cells , Membrane Proteins , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Cell Line , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Humans , Insulin/analysis , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice, Knockout , Signal Transduction/physiology , Wolfram Syndrome
10.
Biophys J ; 117(9): 1764-1777, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31606123

ABSTRACT

Fluorescence fluctuation spectroscopy can be used to measure the aggregation of fluorescently labeled molecules and is typically performed using time series data. Spatial intensity distribution analysis and fluorescence moment image analysis are established tools for measuring molecular brightnesses from single-color images collected with laser scanning microscopes. We have extended these tools for analysis of two-color images to resolve heteromeric interactions between molecules labeled with spectrally distinct chromophores. We call these new methods two-color spatial intensity distribution analysis and two-color spatial cumulant analysis (2c-SpCA). To implement these techniques on a hyperspectral imaging system, we developed a spectral shift filtering technique to remove artifacts due to intrinsic cross talk between detector bins. We determined that 2c-SpCA provides better resolution from samples containing multiple fluorescent species; hence, this technique was carried forward to study images of living cells. We used fluorescent heterodimers labeled with enhanced green fluorescent protein and mApple to quantify the effects of resonance energy transfer and incomplete maturation of mApple on brightness measurements. We show that 2c-SpCA can detect the interaction between two components of trimeric G-protein complexes. Thus, 2c-SpCA presents a robust and computationally expedient means of measuring heteromeric interactions in cellular environments.


Subject(s)
Algorithms , Membrane Proteins/chemistry , Protein Multimerization , Cell Membrane/metabolism , Color , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans
11.
Nat Methods ; 13(7): 557-62, 2016 07.
Article in English | MEDLINE | ID: mdl-27240257

ABSTRACT

The advent of fluorescent proteins (FPs) for genetic labeling of molecules and cells has revolutionized fluorescence microscopy. Genetic manipulations have created a vast array of bright and stable FPs spanning blue to red spectral regions. Common to autofluorescent FPs is their tight ß-barrel structure, which provides the rigidity and chemical environment needed for effectual fluorescence. Despite the common structure, each FP has unique properties. Thus, there is no single 'best' FP for every circumstance, and each FP has advantages and disadvantages. To guide decisions about which FP is right for a given application, we have quantitatively characterized the brightness, photostability, pH stability and monomeric properties of more than 40 FPs to enable straightforward and direct comparison between them. We focus on popular and/or top-performing FPs in each spectral region.


Subject(s)
Luminescent Proteins/analysis , Microscopy, Fluorescence/methods , Recombinant Fusion Proteins/analysis , Spectrometry, Fluorescence/methods , Fluorescence , HeLa Cells , Humans
12.
Diabetes Obes Metab ; 20 Suppl 2: 127-136, 2018 09.
Article in English | MEDLINE | ID: mdl-30230183

ABSTRACT

The islet of Langerhans plays a key role in glucose homeostasis through regulated secretion of the hormones insulin and glucagon. Islet research has focused on the insulin-secreting ß-cells, even though aberrant glucagon secretion from α-cells also contributes to the aetiology of diabetes. Despite its importance, the mechanisms controlling glucagon secretion remain controversial. Proper α-cell function requires the islet milieu, where ß- and δ-cells drive and constrain α-cell dynamics. The response of glucagon to glucose is similar between isolated islets and that measured in vivo, so it appears that the glucose dependence requires only islet-intrinsic factors and not input from blood flow or the nervous system. Elevated intracellular free Ca2+ is needed for α-cell exocytosis, but interpreting Ca2+ data is tricky since it is heterogeneous among α-cells at all physiological glucose levels. Total Ca2+ activity in α-cells increases slightly with glucose, so Ca2+ may serve a permissive, rather than regulatory, role in glucagon secretion. On the other hand, cAMP is a more promising candidate for controlling glucagon secretion and is itself driven by paracrine signalling from ß- and δ-cells. Another pathway, juxtacrine signalling through the α-cell EphA receptors, stimulated by ß-cell ephrin ligands, leads to a tonic inhibition of glucagon secretion. We discuss potential combinations of Ca2+ , cAMP, paracrine and juxtacrine factors in the regulation of glucagon secretion, focusing on recent data in the literature that might unify the field towards a quantitative understanding of α-cell function.


Subject(s)
Calcium/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Blood Glucose/metabolism , Blood Glucose/physiology , Cell Communication/physiology , Cyclic AMP/physiology , Glucagon/antagonists & inhibitors , Humans , Signal Transduction/physiology
13.
Genesis ; 55(9)2017 09.
Article in English | MEDLINE | ID: mdl-28772022

ABSTRACT

During pancreas organogenesis, Neurog3HI endocrine-committing cells are generated from a population of Sox9+ mitotic progenitors with only a low level of Neurog3 transcriptional activity (Neurog3TA.LO ). Low-level Neurog3 protein, in Neurog3TA.LO cells, is required to maintain their mitotic endocrine-lineage-primed status. Herein, we describe a Neurog3-driven FUCCI cell-cycle reporter (Neurog3P2A.FUCCI ) derived from a Neurog3 BAC transgenic reporter that functions as a loxed cassette acceptor (LCA). In cycling Sox9+ Neurog3TA.LO progenitors, the majority of cells in S-G2 -M phases have undetectable levels of Neurog3 with increased expression of endocrine progenitor markers, while those in G1 have low Neurog3 levels with increased expression of endocrine differentiation markers. These findings support a model in which variations in Neurog3 protein levels are coordinated with cell-cycle phase progression in Neurog3TA.LO progenitors with entrance into G1 triggering a concerted effort, beyond increasing Neurog3 levels, to maintain an endocrine-lineage-primed state by initiating expression of the downstream endocrine differentiation program prior to endocrine-commitment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle , Embryonic Stem Cells/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Islets of Langerhans/cytology , Mice , Nerve Tissue Proteins/metabolism
14.
Biophys J ; 111(3): 609-618, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27508444

ABSTRACT

Insulin secretion defects are central to the development of type II diabetes mellitus. Glucose stimulation of insulin secretion has been extensively studied, but its regulation by other stimuli such as incretins and neurotransmitters is not as well understood. We investigated the mechanisms underlying the inhibition of insulin secretion by dopamine, which is synthesized in pancreatic ß-cells from circulating L-dopa. Previous research has shown that this inhibition is mediated primarily by activation of the dopamine receptor D3 subtype (DRD3), even though both DRD2 and DRD3 are expressed in ß-cells. To understand this dichotomy, we investigated the dynamic interactions between the dopamine receptor subtypes and their G-proteins using two-color fluorescence fluctuation spectroscopy (FFS) of mouse MIN6 ß-cells. We show that proper membrane localization of exogenous G-proteins depends on both the Gß and Gγ subunits being overexpressed in the cell. Triple transfections of the dopamine receptor subtype and Gß and Gγ subunits, each labeled with a different-colored fluorescent protein (FP), yielded plasma membrane expression of all three FPs and permitted an FFS evaluation of interactions between the dopamine receptors and the Gßγ complex. Upon dopamine stimulation, we measured a significant decrease in interactions between DRD3 and the Gßγ complex, which is consistent with receptor activation. In contrast, dopamine stimulation did not cause significant changes in the interactions between DRD2 and the Gßγ complex. These results demonstrate that two-color FFS is a powerful tool for measuring dynamic protein interactions in living cells, and show that preferential DRD3 signaling in ß-cells occurs at the level of G-protein release.


Subject(s)
Insulin-Secreting Cells/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Membrane/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Insulin-Secreting Cells/cytology , Mice , Protein Multimerization , Protein Structure, Quaternary , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D3/chemistry , Spectrometry, Fluorescence
15.
Biophys J ; 111(2): 409-417, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27463142

ABSTRACT

The observation of ionic signaling dynamics in intact pancreatic islets has contributed greatly to our understanding of both α- and ß-cell function. Insulin secretion from ß-cells depends on the firing of action potentials and consequent rises of intracellular calcium activity ([Ca(2+)]i). Zinc (Zn(2+)) is cosecreted with insulin, and has been postulated to play a role in cell-to-cell cross talk within an islet, in particular inhibiting glucagon secretion from α-cells. Thus, measuring [Ca(2+)]i and Zn(2+) dynamics from both α- and ß-cells will elucidate mechanisms underlying islet hormone secretion. [Ca(2+)]i and intracellular Zn(2+) can be measured using fluorescent biosensors, but the most efficient sensors have overlapping spectra that complicate their discrimination. Hyperspectral imaging can be used to distinguish signals from multiple fluorophores, but available hyperspectral implementations are either too slow to measure the dynamics of ionic signals or not suitable for thick samples. We have developed a five-dimensional (x,y,z,t,λ) imaging system that leverages a snapshot hyperspectral imaging method, image mapping spectrometry, and light-sheet microscopy. This system provides subsecond temporal resolution from deep within multicellular structures. Using a single excitation wavelength (488 nm) we acquired images from triply labeled samples with two biosensors and a genetically expressing fluorescent protein (spectrally overlapping with one of the biosensors) with high temporal resolution. Measurements of [Ca(2+)]i and Zn(2+) within both α- and ß-cells as a function of glucose concentration show heterogeneous uptake of Zn(2+) into α-cells that correlates to the known heterogeneities in [Ca(2+)]i. These differences in intracellular Zn(2+) among α-cells may contribute to the inhibition in glucagon secretion observed at elevated glucose levels.


Subject(s)
Islets of Langerhans/cytology , Molecular Imaging , Signal Transduction , Animals , Calcium/metabolism , Cell Survival , Intracellular Space/metabolism , Islets of Langerhans/metabolism , Mice , Zinc/metabolism
16.
J Biol Chem ; 289(18): 12435-45, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24627477

ABSTRACT

Glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells is caused by Ca(2+) entry via voltage-dependent Ca(2+) channels. CaMKII is a key mediator and feedback regulator of Ca(2+) signaling in many tissues, but its role in ß-cells is poorly understood, especially in vivo. Here, we report that mice with conditional inhibition of CaMKII in ß-cells show significantly impaired glucose tolerance due to decreased GSIS. Moreover, ß-cell CaMKII inhibition dramatically exacerbates glucose intolerance following exposure to a high fat diet. The impairment of islet GSIS by ß-cell CaMKII inhibition is not accompanied by changes in either glucose metabolism or the activities of KATP and voltage-gated potassium channels. However, glucose-stimulated Ca(2+) entry via voltage-dependent Ca(2+) channels is reduced in islet ß-cells with CaMKII inhibition, as well as in primary wild-type ß-cells treated with a peptide inhibitor of CaMKII. The levels of basal ß-cell cytoplasmic Ca(2+) and of endoplasmic reticulum Ca(2+) stores are also decreased by CaMKII inhibition. In addition, CaMKII inhibition suppresses glucose-stimulated action potential firing frequency. These results reveal that CaMKII is a Ca(2+) sensor with a key role as a feed-forward stimulator of ß-cell Ca(2+) signals that enhance GSIS under physiological and pathological conditions.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Glucose Intolerance/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Action Potentials/drug effects , Animals , Biological Transport/drug effects , Blotting, Western , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Cytoplasm/metabolism , Doxycycline/pharmacology , Endoplasmic Reticulum/metabolism , Glucose/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeostasis/drug effects , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channels/metabolism
17.
Am J Physiol Endocrinol Metab ; 308(12): E1043-55, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25898954

ABSTRACT

Traditional therapies for type 1 diabetes (T1D) involve insulin replacement or islet/pancreas transplantation and have numerous limitations. Our previous work demonstrated the ability of embryonic brown adipose tissue (BAT) transplants to establish normoglycemia without insulin in chemically induced models of insulin-deficient diabetes. The current study sought to extend the technique to an autoimmune-mediated T1D model and document the underlying mechanisms. In nonobese diabetic (NOD) mice, BAT transplants result in complete reversal of T1D associated with rapid and long-lasting euglycemia. In addition, BAT transplants placed prior to the onset of diabetes on NOD mice can prevent or significantly delay the onset of diabetes. As with streptozotocin (STZ)-diabetic models, euglycemia is independent of insulin and strongly correlates with decrease of inflammation and increase of adipokines. Plasma insulin-like growth factor-I (IGF-I) is the first hormone to increase following BAT transplants. Adipose tissue of transplant recipients consistently express IGF-I compared with little or no expression in controls, and plasma IGF-I levels show a direct negative correlation with glucose, glucagon, and inflammatory cytokines. Adipogenic and anti-inflammatory properties of IGF-I may stimulate regeneration of new healthy white adipose tissue, which in turn secretes hypoglycemic adipokines that substitute for insulin. IGF-I can also directly decrease blood glucose through activating insulin receptor. These data demonstrate the potential for insulin-independent reversal of autoimmune-induced T1D with BAT transplants and implicate IGF-I as a likely mediator in the resulting equilibrium.


Subject(s)
Adipose Tissue, Brown/transplantation , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Insulin/metabolism , Adipose Tissue, Brown/embryology , Animals , Blood Glucose/metabolism , Embryo, Mammalian , Female , Fetal Tissue Transplantation , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pregnancy
18.
Am J Physiol Endocrinol Metab ; 308(2): E130-43, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25406263

ABSTRACT

The dysregulation of glucose-inhibited glucagon secretion from the pancreatic islet α-cell is a critical component of diabetes pathology and metabolic disease. We show a previously uncharacterized [Ca(2+)]i-independent mechanism of glucagon suppression in human and murine pancreatic islets whereby cAMP and PKA signaling are decreased. This decrease is driven by the combination of somatostatin, which inhibits adenylyl cyclase production of cAMP via the Gαi subunit of the SSTR2, and insulin, which acts via its receptor to activate phosphodiesterase 3B and degrade cytosolic cAMP. Our data indicate that both somatostatin and insulin signaling are required to suppress cAMP/PKA and glucagon secretion from both human and murine α-cells, and the combination of these two signaling mechanisms is sufficient to reduce glucagon secretion from isolated α-cells as well as islets. Thus, we conclude that somatostatin and insulin together are critical paracrine mediators of glucose-inhibited glucagon secretion and function by lowering cAMP/PKA signaling with increasing glucose.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Somatostatin/pharmacology , Animals , Cyclic AMP/analysis , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Flow Cytometry , Humans , Islets of Langerhans/cytology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Signal Transduction/physiology
19.
Mol Cancer ; 14: 49, 2015 Feb 22.
Article in English | MEDLINE | ID: mdl-25880591

ABSTRACT

BACKGROUND: Non alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases in the United States and worldwide. Our studies have previously shown an increase in metastatic burden in steatotic vs. normal livers using a mouse model of diet induced steatosis. In the present study we aim to identify and evaluate the molecular factors responsible for this increase in tumor burden. METHODS: We assessed changes in expression of a panel of matrix metalloproteinases (MMPs) using qRT-PCR between normal and steatotic livers and validated them with western blot analysis of protein levels. To evaluate the role of MMP13 on tumor development, we utilized a splenic injection model of liver metastasis in Wildtype and Mmp13 deficient mice, using either parental or stable Mmp13 knockdown cell lines. Further, to evaluate changes in the ability of tumor cells to extravasate we utilized whole organ confocal microscopy to identify individual tumor cells relative to the vasculature. MTT, migration and invasion assays were performed to evaluate the role of tumor derived MMP13 on hallmarks of cancer in vitro. RESULTS: We found that MMP13 was significantly upregulated in the steatotic liver both in mice as well as human patients with NAFLD. We showed a decrease in metastatic tumor burden in Mmp13-/- mice compared to wildtype mice, explained in part by a reduction in the number of tumor cells extravasating from the hepatic vasculature in the Mmp13-/- mice compared to wildtype mice. Additionally, loss of tumor derived MMP13 through stable knockdown in tumor cell lines lead to decreased migratory and invasive properties in vitro and metastatic burden in vivo. CONCLUSIONS: This study demonstrates that stromal as well as tumor derived MMP13 contribute to tumor cell extravasation and establishment of metastases in the liver microenvironment.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Matrix Metalloproteinase 13/metabolism , Animals , Cell Movement/genetics , Colorectal Neoplasms/genetics , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression , Humans , Liver Neoplasms/pathology , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Knockout , Neoplasm Invasiveness , Stromal Cells/metabolism
20.
Angew Chem Int Ed Engl ; 54(3): 961-4, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25430909

ABSTRACT

Apoptolidin A has been described among the top 0.1% most-cell-selective cytotoxic agents to be evaluated in the NCI 60 cell line panel. The molecular structure of apoptolidin A consists of a 20-membered macrolide with mono- and disaccharide moieties. In contrast to apoptolidin A, the aglycone (apoptolidinone) shows no cytotoxicity (>10 µM) when evaluated against several tumor cell lines. Apoptolidin H, the C27 deglycosylated analogue of apoptolidin A, displayed sub-micromolar activity against H292 lung carcinoma cells. Selective esterification of apoptolidins A and H with 5-azidopentanoic acid afforded azido-functionalized derivatives of potency equal to that of the parent macrolide. They also underwent strain-promoted alkyne-azido cycloaddition reactions to provide access to fluorescent and biotin-functionalized probes. Microscopy studies demonstrate apoptolidins A and H localize in the mitochondria of H292 human lung carcinoma cells.


Subject(s)
Antineoplastic Agents/chemistry , Fluorescent Dyes/chemistry , Macrolides/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lipid Peroxidation/drug effects , Macrolides/toxicity , Microscopy, Confocal , Pyrones/chemistry , Pyrones/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL