Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Lab Chip ; 24(5): 1327-1350, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38277011

ABSTRACT

Pancreatic islets are metabolically active micron-sized tissues responsible for controlling blood glucose through the secretion of insulin and glucagon. A loss of functional islet mass results in type 1 and 2 diabetes. Islet-on-a-chip devices are powerful microfluidic tools used to trap and study living ex vivo human and murine pancreatic islets and potentially stem cell-derived islet organoids. Devices developed over the past twenty years offer the ability to treat islets with controlled and dynamic microenvironments to mimic in vivo conditions and facilitate diabetes research. In this review, we explore the various islet-on-a-chip devices used to immobilize islets, regulate the microenvironment, and dynamically detect islet metabolism and insulin secretion. We first describe and assess the various methods used to immobilize islets including chambers, dam-walls, and hydrodynamic traps. We subsequently describe the surrounding methods used to create glucose gradients, enhance the reaggregation of dispersed islets, and control the microenvironment of stem cell-derived islet organoids. We focus on the various methods used to measure insulin secretion including capillary electrophoresis, droplet microfluidics, off-chip ELISAs, and on-chip fluorescence anisotropy immunoassays. Additionally, we delve into the various multiparametric readouts (NAD(P)H, Ca2+-activity, and O2-consumption rate) achieved primarily by adopting a microscopy-compatible optical window into the devices. By critical assessment of these advancements, we aim to inspire the development of new devices by the microfluidics community and accelerate the adoption of islet-on-a-chip devices by the wider diabetes research and clinical communities.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Islets of Langerhans , Mice , Humans , Animals , Microfluidics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Lab-On-A-Chip Devices
2.
Cell Rep Methods ; 3(10): 100602, 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37820726

ABSTRACT

First-phase glucose-stimulated insulin secretion is mechanistically linked to type 2 diabetes, yet the underlying metabolism is difficult to discern due to significant islet-to-islet variability. Here, we miniaturize a fluorescence anisotropy immunoassay onto a microfluidic device to measure C-peptide secretion from individual islets as a surrogate for insulin (InsC-chip). This method measures secretion from up to four islets at a time with ∼7 s resolution while providing an optical window for real-time live-cell imaging. Using the InsC-chip, we reveal two glucose-dependent peaks of insulin secretion (i.e., a double peak) within the classically defined 1st phase (<10 min). By combining real-time secretion and live-cell imaging, we show islets transition from glycolytic to oxidative phosphorylation (OxPhos)-driven metabolism at the nadir of the peaks. Overall, these data validate the InsC-chip to measure glucose-stimulated insulin secretion while revealing new dynamics in secretion defined by a shift in glucose metabolism.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Humans , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 2/metabolism , C-Peptide/metabolism , Glucose/pharmacology , Lab-On-A-Chip Devices
3.
Sci Adv ; 9(40): eadi8317, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37792934

ABSTRACT

Several genetically encoded sensors have been developed to study live cell NADPH/NADP+ dynamics, but their use has been predominantly in vitro. Here, we developed an in vivo assay using the Apollo-NADP+ sensor and microfluidic devices to measure endogenous NADPH/NADP+ dynamics in the pancreatic ß cells of live zebrafish embryos. Flux through the pentose phosphate pathway, the main source of NADPH in many cell types, has been reported to be low in ß cells. Thus, it is unclear how these cells compensate to meet NADPH demands. Using our assay, we show that pyruvate cycling is the main source of NADP+ reduction in ß cells, with contributions from folate cycling after acute electrical activation. INS1E ß cells also showed a stress-induced increase in folate cycling and further suggested that this cycling requires both increased glycolytic intermediates and cytosolic NAD+. Overall, we show in vivo application of the Apollo-NADP+ sensor and reveal that ß cells are capable of adapting NADPH/NADP+ redox during stress.


Subject(s)
Insulin-Secreting Cells , Animals , NADP/metabolism , Zebrafish/metabolism , Oxidation-Reduction , Folic Acid/metabolism
4.
ACS Sens ; 7(11): 3308-3317, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36269889

ABSTRACT

NADPH/NADP+ redox state supports numerous reactions related to cell growth and survival; yet the full impact is difficult to appreciate due to organelle compartmentalization of NADPH and NADP+. To study glucose-stimulated NADPH production in pancreatic beta-cell organelles, we targeted the Apollo-NADP+ sensor by first selecting the most pH-stable version of the single-color sensor. We subsequently targeted mTurquoise2-Apollo-NADP+ to various organelles and confirmed activity in the cytoplasm, mitochondrial matrix, nucleus, and peroxisome. Finally, we measured the glucose- and glutamine-stimulated NADPH responses by single- and dual-color imaging of the targeted sensors. Overall, we developed multiple organelle-targeted Apollo-NADP+ sensors to reveal the prominent role of beta-cell mitochondria in determining NADPH production in the cytoplasm, nucleus, and peroxisome.


Subject(s)
Insulin-Secreting Cells , NADP/metabolism , Insulin-Secreting Cells/metabolism , Oxidation-Reduction , Glucose/metabolism , Mitochondria/metabolism
5.
Biofabrication ; 14(4)2022 07 19.
Article in English | MEDLINE | ID: mdl-35793653

ABSTRACT

Precision-cut-tissues (PCTs), which preserve many aspects of a tissue's microenvironment, are typically imaged using conventional sample dishes and chambers. These can require large amounts of reagent and, when used for flow-through experiments, the shear forces applied on the tissues are often ill-defined. Their physical design also makes it difficult to image large volumes and repetitively image smaller regions of interest in the living slice. We report here on the design of a versatile microfluidic device capable of holding mouse or human pancreas PCTs for 3D fluorescence imaging using confocal and selective plane illumination microscopy (SPIM). Our design positions PCTs within a 5 × 5 mm × 140µm deep chamber fitted with 150µm tall channels to facilitate media exchange. Shear stress in the device is localized to small regions on the surface of the tissue and can be easily controlled. This design allows for media exchange at flowrates ∼10-fold lower than those required for conventional chambers. Finally, this design allows for imaging the same immunofluorescently labeled PCT with high resolution on a confocal and with large field of view on a SPIM, without adversely affecting image quality.


Subject(s)
Imaging, Three-Dimensional , Lab-On-A-Chip Devices , Animals , Humans , Imaging, Three-Dimensional/methods , Mice , Microscopy, Fluorescence/methods , Optical Imaging
6.
Sci Rep ; 12(1): 6110, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35414066

ABSTRACT

We previously showed that pancreatic beta-cells plated on laminin matrix express reduced levels of FGFR1, a receptor linked to beta-cell metabolism and differentiation. Due to recent evidence that adult beta-cells also express FGFR5, a co-receptor for FGFR1, we now aim to determine the effect of laminin on FGFR5 expression and consequent effects on beta-cell metabolism. Using a genetically encoded sensor for NADPH/NADP+ redox state (Apollo-NADP+), we show overexpression of FGFR5 enhances glucose-stimulated NADPH metabolism in beta-cell lines as well as mouse and human beta-cells. This enhanced response was accompanied by increased insulin secretion as well as increased expression of transcripts for glycolytic enzymes (Glucokinase/GCK, PKM2) and the functional maturity marker Urocortin 3 (UCN3). Culturing beta-cells on laminin matrix also stimulated upregulation of endogenous FGFR5 expression, and similarly enhanced beta-cell glucose-stimulated NADPH-metabolism as well as GCK and PKM2 transcript expression. The metabolism and transcript responses triggered by laminin were disrupted by R5ΔC, a truncated receptor isoform that inhibits the FGFR5/FGFR1 signaling complex. Collectively these data reveal that beta-cells respond to laminin by increasing FGFR5 expression to enhance beta-cell glucose metabolism.


Subject(s)
Insulin-Secreting Cells , Laminin , Animals , Glucokinase/metabolism , Glucose/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Laminin/metabolism , Mice , NADP/metabolism
7.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-34027899

ABSTRACT

GWAS have shown that the common R325W variant of SLC30A8 (ZnT8) increases the risk of type 2 diabetes (T2D). However, ZnT8 haploinsufficiency is protective against T2D in humans, counterintuitive to earlier work in humans and mouse models. Therefore, whether decreasing ZnT8 activity is beneficial or detrimental to ß cell function, especially under conditions of metabolic stress, remains unknown. In order to examine whether the existence of human islet amyloid polypeptide (hIAPP), a coresident of the insulin granule, affects the role of ZnT8 in regulating ß cell function, hIAPP-expressing transgenics were generated with reduced ZnT8 (ZnT8B+/- hIAPP) or null ZnT8 (ZnT8B-/- hIAPP) expression specifically in ß cells. We showed that ZnT8B-/- hIAPP mice on a high-fat diet had intensified amyloid deposition and further impaired glucose tolerance and insulin secretion compared with control, ZnT8B-/-, and hIAPP mice. This can in part be attributed to impaired glucose sensing and islet cell synchronicity. Importantly, ZnT8B+/- hIAPP mice were also glucose intolerant and had reduced insulin secretion and increased amyloid aggregation compared with controls. These data suggest that loss of or reduced ZnT8 activity in ß cells heightened the toxicity induced by hIAPP, leading to impaired ß cell function and glucose homeostasis associated with metabolic stress.


Subject(s)
Amyloidosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Zinc Transporter 8 , Animals , Disease Models, Animal , Humans , Islet Amyloid Polypeptide/genetics , Male , Mice , Mice, Transgenic , Zinc Transporter 8/genetics , Zinc Transporter 8/metabolism
8.
Diabetes ; 70(8): 1717-1728, 2021 08.
Article in English | MEDLINE | ID: mdl-34039628

ABSTRACT

The defining feature of pancreatic islet ß-cell function is the precise coordination of changes in blood glucose levels with insulin secretion to regulate systemic glucose homeostasis. While ATP has long been heralded as a critical metabolic coupling factor to trigger insulin release, glucose-derived metabolites have been suggested to further amplify fuel-stimulated insulin secretion. The mitochondrial export of citrate and isocitrate through the citrate-isocitrate carrier (CIC) has been suggested to initiate a key pathway that amplifies glucose-stimulated insulin secretion, though the physiological significance of ß-cell CIC-to-glucose homeostasis has not been established. Here, we generated constitutive and adult CIC ß-cell knockout (KO) mice and demonstrate that these animals have normal glucose tolerance, similar responses to diet-induced obesity, and identical insulin secretion responses to various fuel secretagogues. Glucose-stimulated NADPH production was impaired in ß-cell CIC KO islets, whereas glutathione reduction was retained. Furthermore, suppression of the downstream enzyme cytosolic isocitrate dehydrogenase (Idh1) inhibited insulin secretion in wild-type islets but failed to impact ß-cell function in ß-cell CIC KO islets. Our data demonstrate that the mitochondrial CIC is not required for glucose-stimulated insulin secretion and that additional complexities exist for the role of Idh1 and NADPH in the regulation of ß-cell function.


Subject(s)
Citric Acid/metabolism , Glucose/pharmacology , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Isocitrates/metabolism , Mitochondria/metabolism , Animals , Cytosol/metabolism , Homeostasis/drug effects , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout , Mitochondria/drug effects
9.
Mol Metab ; 47: 101185, 2021 05.
Article in English | MEDLINE | ID: mdl-33561544

ABSTRACT

OBJECTIVE: Autophagy is a physiological self-eating process that can promote cell survival or activate cell death in eukaryotic cells. In skeletal muscle, it is important for maintaining muscle mass and function that is critical to sustain mobility and regulate metabolism. The UV radiation resistance-associated gene (UVRAG) regulates the early stages of autophagy and autophagosome maturation and plays a key role in endosomal trafficking. This study investigated the essential in vivo role of UVRAG in skeletal muscle biology. METHODS: To determine the role of UVRAG in skeletal muscle in vivo, we generated muscle-specific UVRAG knockout mice using the Cre-loxP system driven by Myf6 promoter that is exclusively expressed in skeletal muscle. Myf6-Cre+ UVRAGfl/fl (M-UVRAG-/-) mice were compared to littermate Myf6-Cre+ UVRAG+/+ (M-UVRAG+/+) controls under basal conditions on a normal chow diet. Body composition, muscle function, and mitochondria morphology were assessed in muscles of the WT and KO mice at 24 weeks of age. RESULTS: M-UVRAG-/- mice developed accelerated sarcopenia and impaired muscle function compared to M-UVRAG+/+ littermates at 24 weeks of age. Interestingly, these mice displayed improved glucose tolerance and increased energy expenditure likely related to upregulated Fgf21, a marker of muscle dysfunction. Skeletal muscle of the M-UVRAG-/- mice showed altered mitochondrial morphology with increased mitochondrial fission and EGFR accumulation reflecting defects in endosomal trafficking. To determine whether increased EGFR signaling had a causal role in muscle dysfunction, the mice were treated with an EGFR inhibitor, gefitinib, which partially restored markers of muscle and mitochondrial deregulation. Conversely, constitutively active EGFR transgenic expression in UVRAG-deficient muscle led to further detrimental effects with non-overlapping distinct defects in muscle function, with EGFR activation affecting the muscle fiber type whereas UVRAG deficiency impaired mitochondrial homeostasis. CONCLUSIONS: Our results show that both UVRAG and EGFR signaling are critical for maintaining muscle mass and function with distinct mechanisms in the differentiation pathway.


Subject(s)
ErbB Receptors/metabolism , Homeostasis , Muscle, Skeletal/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Autophagy , Endosomes/metabolism , ErbB Receptors/genetics , Female , Fibroblast Growth Factors/metabolism , Male , Mice , Mice, Knockout , Mitochondrial Dynamics , Transcriptome , Tumor Suppressor Proteins/genetics , Ultraviolet Rays
10.
APL Bioeng ; 5(1): 016101, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33415313

ABSTRACT

Deep learning provides an opportunity to automatically segment and extract cellular features from high-throughput microscopy images. Many labeling strategies have been developed for this purpose, ranging from the use of fluorescent markers to label-free approaches. However, differences in the channels available to each respective training dataset make it difficult to directly compare the effectiveness of these strategies across studies. Here, we explore training models using subimage stacks composed of channels sampled from larger, "hyper-labeled," image stacks. This allows us to directly compare a variety of labeling strategies and training approaches on identical cells. This approach revealed that fluorescence-based strategies generally provide higher segmentation accuracies but were less accurate than label-free models when labeling was inconsistent. The relative strengths of label and label-free techniques could be combined through the use of merging fluorescence channels and using out-of-focus brightfield images. Beyond comparing labeling strategies, using subimage stacks for training was also found to provide a method of simulating a wide range of labeling conditions, increasing the ability of the final model to accommodate a greater range of candidate cell labeling strategies.

11.
Adv Mater ; 32(24): e1906274, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32383233

ABSTRACT

Nanoparticles are commonly administered through systemic injection, which exposes them to the dynamic environment of the bloodstream. Injected nanoparticles travel within the blood and experience a wide range of flow velocities that induce varying shear rates to the blood vessels. Endothelial cells line these vessels, and have been shown to uptake nanoparticles during circulation, but it is difficult to characterize the flow-dependence of this interaction in vivo. Here, a microfluidic system is developed to control the flow rates of nanoparticles as they interact with endothelial cells. Gold nanoparticle uptake into endothelial cells is quantified at varying flow rates, and it is found that increased flow rates lead to decreased nanoparticle uptake. Endothelial cells respond to increased flow shear with decreased ability to uptake the nanoparticles. If cells are sheared the same way, nanoparticle uptake decreases as their flow velocity increases. Modifying nanoparticle surfaces with endothelial-cell-binding ligands partially restores uptake to nonflow levels, suggesting that functionalizing nanoparticles to bind to endothelial cells enables nanoparticles to resist flow effects. In the future, this microfluidic system can be used to test other nanoparticle-endothelial cell interactions under flow. The results of these studies can guide the engineering of nanoparticles for in vivo medical applications.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Blood Circulation , Human Umbilical Vein Endothelial Cells/metabolism , Nanoparticles , Biocompatible Materials/administration & dosage , Biological Transport , Cell Communication , Human Umbilical Vein Endothelial Cells/cytology , Humans , Injections , Kinetics
12.
PLoS One ; 14(10): e0222424, 2019.
Article in English | MEDLINE | ID: mdl-31600313

ABSTRACT

BACKGROUND: Pancreatic islets are heavily vascularized in vivo yet lose this vasculature after only a few days in culture. Determining how to maintain islet vascularity in culture could lead to better outcomes in transplanting this tissue for the treatment of type 1 diabetes as well as provide insight into the complex communication between beta-cells and endothelial cells (ECs). We previously showed that islet ECs die in part due to limited diffusion of serum albumin into the tissue. We now aim to determine the impact of hypoxia on islet vascularization. METHODS: We induced hypoxia in cultured mouse islets using the hypoxia mimetic cobalt chloride (100 µM CoCl2). We measured the impact on islet metabolism (two-photon NAD(P)H and Rh123 imaging) and function (insulin secretion and survival). We also measured the impact on hypoxia related transcripts (HIF-1α, VEGF-A, PDK-1, LDHA, COX4) and confirmed increased VEGF-A expression and secretion. Finally, we measured the vascularization of islets in static and flowing culture using PECAM-1 immunofluorescence. RESULTS: CoCl2 did not induce significant changes in beta cell metabolism (NAD(P)H and Rh123), insulin secretion, and survival. Consistent with hypoxia induction, CoCl2 stimulated HIF-1α, PDK-1, and LDHA transcripts and also stimulated VEGF expression and secretion. We observed a modest switch to the less oxidative isoform of COX4 (isoform 1 to 2) and this switch was noted in the glucose-stimulated cytoplasmic NAD(P)H responses. EC morphology and survival were greater in CoCl2 treated islets compared to exogenous VEGF-A in both static (dish) and microfluidic flow culture. CONCLUSIONS: Hypoxia induction using CoCl2 had a positive effect on islet EC morphology and survival with limited impact on beta-cell metabolism, function, and survival. The EC response appears to be due to endogenous production and secretion of angiogenic factors (e.g. VEGF-A), and mechanistically independent from survival induced by serum albumin.


Subject(s)
Cell Communication/genetics , Endothelial Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Animals , Cell Communication/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cobalt/pharmacology , Electron Transport Complex IV/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Insulin/genetics , Insulin/metabolism , Insulin Secretion/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Vascular Endothelial Growth Factor A/genetics
13.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217817

ABSTRACT

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Subject(s)
Cytokines/immunology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Animals , Diabetes Mellitus/immunology , Dimerization , Female , Fibroblast Growth Factor 2/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 5/chemistry , Receptor, Fibroblast Growth Factor, Type 5/immunology , Up-Regulation
14.
ACS Appl Mater Interfaces ; 9(24): 20435-20443, 2017 Jun 21.
Article in English | MEDLINE | ID: mdl-28548481

ABSTRACT

Isolating subpopulations of heterogeneous cancer cells is an important capability for the meaningful characterization of circulating tumor cells at different stages of tumor progression and during the epithelial-to-mesenchymal transition. Here, we present a microfluidic device that can separate phenotypically distinct subpopulations of cancer cells. Magnetic nanoparticles coated with antibodies against the epithelial cell adhesion molecule (EpCAM) are used to separate breast cancer cells in the microfluidic platform. Cells are sorted into different zones on the basis of the levels of EpCAM expression, which enables the detection of cells that are losing epithelial character and becoming more mesenchymal. The phenotypic properties of the isolated cells with low and high EpCAM are then assessed using matrix-coated surfaces for collagen uptake analysis, and an NAD(P)H assay that assesses metabolic activity. We show that low-EpCAM expressing cells have higher collagen uptake and higher folate-induced NAD(P)H responses compared to those of high-EpCAM expressing cells. In addition, we tested SKBR3 cancer cells undergoing chemically induced hypoxia. The induced cells have reduced expression of EpCAM, and we find that these cells have higher collagen uptake and NAD(P)H metabolism relative to noninduced cells. This work demonstrates that nanoparticle-mediated binning facilitates the isolation of functionally distinct cell subpopulations and allows surface marker expression to be associated with invasiveness, including collagen uptake and metabolic activity.


Subject(s)
Nanoparticles , Antigens, Neoplasm , Cell Adhesion Molecules , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Humans , Neoplastic Cells, Circulating
15.
Cell Rep ; 18(10): 2301-2309, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28273447

ABSTRACT

Mitochondria undergo dynamic changes to maintain function in eukaryotic cells. Insulin action in parallel regulates glucose homeostasis, but whether specific changes in mitochondrial dynamics alter insulin action and glucose homeostasis remains elusive. Here, we report that high-fat feeding in rodents incurred adaptive dynamic changes in mitochondria through an increase in mitochondrial fission in parallel to an activation of dynamin-related protein 1 (Drp1) in the dorsal vagal complex (DVC) of the brain. Direct inhibition of Drp1 negated high-fat-feeding-induced mitochondrial fission, endoplasmic reticulum (ER) stress, and insulin resistance in the DVC and subsequently restored hepatic glucose production regulation. Conversely, molecular activation of DVC Drp1 in healthy rodents was sufficient to induce DVC mitochondrial fission, ER stress, and insulin resistance. Together, these data illustrate that Drp1-dependent mitochondrial fission changes in the DVC regulate insulin action and suggest that targeting the Drp1-mitochondrial-dependent pathway in the brain may have therapeutic potential in insulin resistance.


Subject(s)
Brain/metabolism , Dynamins/metabolism , Insulin/metabolism , Mitochondrial Dynamics , Animals , Diet, High-Fat , Endoplasmic Reticulum Stress , HEK293 Cells , Humans , Insulin Resistance , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats, Sprague-Dawley
16.
Proc Natl Acad Sci U S A ; 113(52): 14915-14920, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27956625

ABSTRACT

On-chip imaging of intact three-dimensional tissues within microfluidic devices is fundamentally hindered by intratissue optical scattering, which impedes their use as tissue models for high-throughput screening assays. Here, we engineered a microfluidic system that preserves and converts tissues into optically transparent structures in less than 1 d, which is 20× faster than current passive clearing approaches. Accelerated clearing was achieved because the microfluidic system enhanced the exchange of interstitial fluids by 567-fold, which increased the rate of removal of optically scattering lipid molecules from the cross-linked tissue. Our enhanced clearing process allowed us to fluorescently image and map the segregation and compartmentalization of different cells during the formation of tumor spheroids, and to track the degradation of vasculature over time within extracted murine pancreatic islets in static culture, which may have implications on the efficacy of beta-cell transplantation treatments for type 1 diabetes. We further developed an image analysis algorithm that automates the analysis of the vasculature connectivity, volume, and cellular spatial distribution of the intact tissue. Our technique allows whole tissue analysis in microfluidic systems, and has implications in the development of organ-on-a-chip systems, high-throughput drug screening devices, and in regenerative medicine.

17.
J Am Chem Soc ; 138(36): 11583-98, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27494760

ABSTRACT

G protein-coupled receptors constitute the largest family of transmembrane signaling proteins and the largest pool of drug targets, yet their mechanism of action remains obscure. That uncertainty relates to unresolved questions regarding the supramolecular nature of the signaling complex formed by receptor and G protein. We therefore have characterized the oligomeric status of eGFP-tagged M2 muscarinic receptor (M2R) and Gi1 by single-particle photobleaching of immobilized complexes. The method was calibrated with multiplexed controls comprising 1-4 copies of fused eGFP. The photobleaching patterns of eGFP-M2R were indicative of a tetramer and unaffected by muscarinic ligands; those of eGFP-Gi1 were indicative of a hexamer and unaffected by GTPγS. A complex of M2R and Gi1 was tetrameric in both, and activation by a full agonist plus GTPγS reduced the oligomeric size of Gi1 without affecting that of the receptor. A similar reduction was observed upon activation of eGFP-Gαi1 by the receptor-mimic mastoparan plus GTPγS, and constitutively active eGFP-Gαi1 was predominantly dimeric. The oligomeric nature of Gi1 in live CHO cells was demonstrated by means of Förster resonance energy transfer and dual-color fluorescence correlation spectroscopy in studies with eGFP- and mCherry-labeled Gαi1; stochastic FRET was ruled out by means of non-interacting pairs. These results suggest that the complex between M2R and holo-Gi1 is an octamer comprising four copies of each, and that activation is accompanied by a decrease in the oligomeric size of Gi1. The structural feasibility of such a complex was demonstrated in molecular dynamics simulations.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Molecular Dynamics Simulation , Receptor, Muscarinic M2/chemistry , Animals , CHO Cells , Cricetulus , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Protein Multimerization , Protein Structure, Quaternary , Receptor, Muscarinic M2/metabolism
18.
Lab Chip ; 16(15): 2921-34, 2016 08 07.
Article in English | MEDLINE | ID: mdl-27378588

ABSTRACT

Tissues are challenging to genetically manipulate due to limited penetration of viral particles resulting in low transduction efficiency. We are particularly interested in expressing genetically-encoded sensors in ex vivo pancreatic islets to measure glucose-stimulated metabolism, however poor viral penetration biases these measurements to only a subset of cells at the periphery. To increase mass transfer of viral particles, we designed a microfluidic device that holds islets in parallel hydrodynamic traps connected by an expanding by-pass channel. We modeled viral particle flow into the tissue using fluorescently-labelled gold nanoparticles of varying sizes and showed a penetration threshold of only ∼5 nm. To increase this threshold, we used EDTA to transiently reduce cell-cell adhesion and expand intercellular space. Ultimately, a combination of media flow and ETDA treatment significantly increased adenoviral transduction to the core of the islet. As proof-of-principle, we used this protocol to transduce an ER-targeted redox sensitive sensor (eroGFP), and revealed significantly greater ER redox capacity at core islet cells. Overall, these data demonstrate a robust method to enhance transduction efficiency of islets, and potentially other tissues, by using a combination of microfluidic flow and transient tissue expansion.


Subject(s)
Adenoviridae/physiology , Endoplasmic Reticulum Stress , Gene Transfer Techniques/instrumentation , Islets of Langerhans/virology , Lab-On-A-Chip Devices , Models, Biological , Virion/physiology , Animals , Calcium Chelating Agents/pharmacology , Cell Survival/drug effects , Dithiothreitol/toxicity , Endoplasmic Reticulum Stress/drug effects , Equipment Design , Feasibility Studies , Gold/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Metal Nanoparticles/chemistry , Mice, Inbred C57BL , Proof of Concept Study , Recombinant Fusion Proteins/metabolism , Reducing Agents/toxicity , Tissue Culture Techniques
19.
Elife ; 52016 05 06.
Article in English | MEDLINE | ID: mdl-27151542

ABSTRACT

The M2 muscarinic receptor is the prototypic model of allostery in GPCRs, yet the molecular and the supramolecular determinants of such effects are unknown. Monomers and oligomers of the M2 muscarinic receptor therefore have been compared to identify those allosteric properties that are gained in oligomers. Allosteric interactions were monitored by means of a FRET-based sensor of conformation at the allosteric site and in pharmacological assays involving mutants engineered to preclude intramolecular effects. Electrostatic, steric, and conformational determinants of allostery at the atomic level were examined in molecular dynamics simulations. Allosteric effects in monomers were exclusively negative and derived primarily from intramolecular electrostatic repulsion between the allosteric and orthosteric ligands. Allosteric effects in oligomers could be positive or negative, depending upon the allosteric-orthosteric pair, and they arose from interactions within and between the constituent protomers. The complex behavior of oligomers is characteristic of muscarinic receptors in myocardial preparations.


Subject(s)
Allosteric Regulation/genetics , Molecular Conformation , Receptor, Muscarinic M2/metabolism , Receptors, G-Protein-Coupled/metabolism , Allosteric Site/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Multimerization/genetics , Receptor, Muscarinic M2/chemistry , Receptor, Muscarinic M2/genetics , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics
20.
Nat Methods ; 13(4): 352-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878383

ABSTRACT

NADPH-dependent antioxidant pathways have a critical role in scavenging hydrogen peroxide (H2O2) produced by oxidative phosphorylation. Inadequate scavenging results in H2O2 accumulation and can cause disease. To measure NADPH/NADP(+) redox states, we explored genetically encoded sensors based on steady-state fluorescence anisotropy due to FRET (fluorescence resonance energy transfer) between homologous fluorescent proteins (homoFRET); we refer to these sensors as Apollo sensors. We created an Apollo sensor for NADP(+) (Apollo-NADP(+)) that exploits NADP(+)-dependent homodimerization of enzymatically inactive glucose-6-phosphate dehydrogenase (G6PD). This sensor is reversible, responsive to glucose-stimulated metabolism and spectrally tunable for compatibility with many other sensors. We used Apollo-NADP(+) to study beta cells responding to oxidative stress and demonstrated that NADPH is significantly depleted before H2O2 accumulation by imaging a Cerulean-tagged version of Apollo-NADP(+) with the H2O2 sensor HyPer.


Subject(s)
Biosensing Techniques/methods , Glucosephosphate Dehydrogenase/metabolism , Insulin-Secreting Cells/metabolism , NADP/chemistry , Cells, Cultured , Fluorescence Polarization/methods , Fluorescence Resonance Energy Transfer , Glucosephosphate Dehydrogenase/chemistry , Glucosephosphate Dehydrogenase/genetics , Humans , Hydrogen Peroxide/metabolism , Image Processing, Computer-Assisted , NADP/metabolism , Oxidants/metabolism , Oxidative Stress , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...