Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Matthew J. Merrins is active.

Publication


Featured researches published by Matthew J. Merrins.


Journal of Biological Chemistry | 2013

Direct Measurements of Oscillatory Glycolysis in Pancreatic Islet β-cells using Novel Fluorescence Resonance Energy Transfer(FRET) Biosensors for Pyruvate Kinase M2 Activity

Matthew J. Merrins; Aaron R. Van Dyke; Anna K. Mapp; Mark A. Rizzo; Leslie S. Satin

Background: Pyruvate kinase M2 controls glycolytic efflux. Results: Novel FRET sensors report PKM2 structural changes in response to activation by metabolites and phosphorylation events in pancreatic β-cells. Conclusion: PKM2 activity is oscillatory and synchronized between islet β-cells. Significance: This approach is broadly applicable for measuring pyruvate kinase M2 activity in living cells. Pulses of insulin released from pancreatic β-cells maintain blood glucose in a narrow range, although the source of these pulses is unclear. We and others have proposed that positive feedback mediated by the glycolytic enzyme phosphofructokinase-1 (PFK1) enables β-cells to generate metabolic oscillations via autocatalytic activation by its product fructose 1,6-bisphosphate (FBP). Although much indirect evidence has accumulated in favor of this hypothesis, a direct measurement of oscillating glycolytic intermediates has been lacking. To probe glycolysis directly, we engineered a family of inter- and intramolecular FRET biosensors based on the glycolytic enzyme pyruvate kinase M2 (PKAR; pyruvate kinase activity reporter), which multimerizes and is activated upon binding FBP. When introduced into Min6 β-cells, PKAR FRET efficiency increased rapidly in response to glucose. Importantly, however, metabolites entering downstream of PFK1 (glyceraldehyde, pyruvate, and ketoisocaproate) failed to activate PKAR, consistent with sensor activation by FBP; the dependence of PKAR on FBP was further confirmed using purified sensor in vitro. Using a novel imaging modality for monitoring mitochondrial flavin fluorescence in mouse islets, we show that slow oscillations in mitochondrial redox potential stimulated by 10 mm glucose are in phase with glycolytic efflux through PKM2, measured simultaneously from neighboring islet β-cells expressing PKAR. These results indicate that PKM2 activity in β-cells is oscillatory and are consistent with pulsatile PFK1 being the mediator of slow glycolytic oscillations.


Biophysical Journal | 2010

Metabolic Oscillations in Pancreatic Islets Depend on the Intracellular Ca2+ Level but Not Ca2+ Oscillations

Matthew J. Merrins; Bernard Fendler; Min Zhang; Arthur Sherman; Richard Bertram; Leslie S. Satin

Plasma insulin is pulsatile and reflects oscillatory insulin secretion from pancreatic islets. Although both islet Ca(2+) and metabolism oscillate, there is disagreement over their interrelationship, and whether they can be dissociated. In some models of islet oscillations, Ca(2+) must oscillate for metabolic oscillations to occur, whereas in others metabolic oscillations can occur without Ca(2+) oscillations. We used NAD(P)H fluorescence to assay oscillatory metabolism in mouse islets stimulated by 11.1 mM glucose. After abolishing Ca(2+) oscillations with 200 microM diazoxide, we observed that oscillations in NAD(P)H persisted in 34% of islets (n = 101). In the remainder of the islets (66%) both Ca(2+) and NAD(P)H oscillations were eliminated by diazoxide. However, in most of these islets NAD(P)H oscillations could be restored and amplified by raising extracellular KCl, which elevated the intracellular Ca(2+) level but did not restore Ca(2+) oscillations. Comparatively, we examined islets from ATP-sensitive K(+) (K(ATP)) channel-deficient SUR1(-/-) mice. Again NAD(P)H oscillations were evident even though Ca(2+) and membrane potential oscillations were abolished. These observations are predicted by the dual oscillator model, in which intrinsic metabolic oscillations and Ca(2+) feedback both contribute to the oscillatory islet behavior, but argue against other models that depend on Ca(2+) oscillations for metabolic oscillations to occur.


The Journal of Physiology | 2008

Kinetics of Rab27a-dependent actions on vesicle docking and priming in pancreatic β-cells

Matthew J. Merrins; Edward L. Stuenkel

The small GTPase Rab27a, along with the isoforms of Rab3, is present on insulin secretory granules and has been implicated in regulation of Ca2+‐triggered exocytosis. We have used membrane capacitance measurements to define the role of Rab27a in regulating the size and refilling of distinct pools of insulin granules by comparison of evoked secretory responses from Rab27a‐null ashen and strain‐matched wild‐type control pancreatic β‐cells. We find that ashenβ‐cells display a kinetic defect in refilling of readily releasable and immediately releasable vesicle pools (RRP and IRP, respectively) in response to depolarization‐evoked Ca2+ influx. The deficit in IRP refilling was not observed in the presence of stimulatory glucose concentrations (16.7 mm), though incomplete refilling of the RRP persisted. Comparatively, β‐cells from Rab3a−/− mice exhibited complete refilling of the IRP and RRP, demonstrating that Rab27a and Rab3a exert distinct roles in the insulin granule secretory pathway. Further, depletion of the RRP in ashenβ‐cells was twofold faster than that of control β‐cells. These deficits in refilling and exocytotic rate in ashenβ‐cells were absent when cAMP‐regulatory pathways were activated. Elevated cAMP increased the RRP pool size, and complete refilling of the RRP occurred in ashenβ‐cells; responses were comparable to wild‐type controls. These effects of cAMP were largely eliminated by Rp‐cAMP inhibition of PKA, indicating that PKA acts on vesicle priming downstream or via pathways independent of Rab27a. In summary, Rab27a exerts dual roles in glucose‐mediated insulin granule exocytosis, facilitating refilling of releasable granule pools while also limiting the rate of release from these pools.


Journal of Clinical Investigation | 2014

Maternal diet–induced microRNAs and mTOR underlie β cell dysfunction in offspring

Emilyn U. Alejandro; Brigid Gregg; Taylor Wallen; Doga Kumusoglu; Daniel L. Meister; Angela Chen; Matthew J. Merrins; Leslie S. Satin; Ming Liu; Peter Arvan; Ernesto Bernal-Mizrachi

A maternal diet that is low in protein increases the susceptibility of offspring to type 2 diabetes by inducing long-term alterations in β cell mass and function. Nutrients and growth factor signaling converge through mTOR, suggesting that this pathway participates in β cell programming during fetal development. Here, we revealed that newborns of dams exposed to low-protein diet (LP0.5) throughout pregnancy exhibited decreased insulin levels, a lower β cell fraction, and reduced mTOR signaling. Adult offspring of LP0.5-exposed mothers exhibited glucose intolerance as a result of an insulin secretory defect and not β cell mass reduction. The β cell insulin secretory defect was distal to glucose-dependent Ca2+ influx and resulted from reduced proinsulin biosynthesis and insulin content. Islets from offspring of LP0.5-fed dams exhibited reduced mTOR and increased expression of a subset of microRNAs, and blockade of microRNA-199a-3p and -342 in these islets restored mTOR and insulin secretion to normal. Finally, transient β cell activation of mTORC1 signaling in offspring during the last week of pregnancy of mothers fed a LP0.5 rescued the defect in the neonatal β cell fraction and metabolic abnormalities in the adult. Together, these findings indicate that a maternal low-protein diet alters microRNA and mTOR expression in the offspring, influencing insulin secretion and glucose homeostasis.


PLOS ONE | 2009

Glucose metabolism, islet architecture, and genetic homogeneity in imprinting of [Ca2+](i) and insulin rhythms in mouse islets.

Craig S. Nunemaker; John F. Dishinger; Stacey B. Dula; Runpei Wu; Matthew J. Merrins; Kendra R. Reid; Arthur Sherman; Robert T. Kennedy; Leslie S. Satin

We reported previously that islets isolated from individual, outbred Swiss-Webster mice displayed oscillations in intracellular calcium ([Ca2+]i) that varied little between islets of a single mouse but considerably between mice, a phenomenon we termed “islet imprinting.” We have now confirmed and extended these findings in several respects. First, imprinting occurs in both inbred (C57BL/6J) as well as outbred mouse strains (Swiss-Webster; CD1). Second, imprinting was observed in NAD(P)H oscillations, indicating a metabolic component. Further, short-term exposure to a glucose-free solution, which transiently silenced [Ca2+]i oscillations, reset the oscillatory patterns to a higher frequency. This suggests a key role for glucose metabolism in maintaining imprinting, as transiently suppressing the oscillations with diazoxide, a KATP-channel opener that blocks [Ca2+]i influx downstream of glucose metabolism, did not change the imprinted patterns. Third, imprinting was not as readily observed at the level of single beta cells, as the [Ca2+]i oscillations of single cells isolated from imprinted islets exhibited highly variable, and typically slower [Ca2+]i oscillations. Lastly, to test whether the imprinted [Ca2+]i patterns were of functional significance, a novel microchip platform was used to monitor insulin release from multiple islets in real time. Insulin release patterns correlated closely with [Ca2+]i oscillations and showed significant mouse-to-mouse differences, indicating imprinting. These results indicate that islet imprinting is a general feature of islets and is likely to be of physiological significance. While islet imprinting did not depend on the genetic background of the mice, glucose metabolism and intact islet architecture may be important for the imprinting phenomenon.


PLOS ONE | 2012

Phosphofructo-2-kinase/Fructose-2,6-bisphosphatase Modulates Oscillations of Pancreatic Islet Metabolism

Matthew J. Merrins; Richard Bertram; Arthur Sherman; Leslie S. Satin

Pulses of insulin from pancreatic beta-cells help maintain blood glucose in a narrow range, although the source of these pulses is unclear. It has been proposed that a positive feedback circuit exists within the glycolytic pathway, the autocatalytic activation of phosphofructokinase-1 (PFK1), which endows pancreatic beta-cells with the ability to generate oscillations in metabolism. Flux through PFK1 is controlled by the bifunctional enzyme PFK2/FBPase2 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) in two ways: via (1) production/degradation of fructose-2,6-bisphosphate (Fru2,6-BP), a potent allosteric activator of PFK1, as well as (2) direct activation of glucokinase due to a protein-protein interaction. In this study, we used a combination of live-cell imaging and mathematical modeling to examine the effects of inducibly-expressed PFK2/FBPase2 mutants on glucose-induced Ca2+ pulsatility in mouse islets. Irrespective of the ability to bind glucokinase, mutants of PFK2/FBPase2 that increased the kinase:phosphatase ratio reduced the period and amplitude of Ca2+ oscillations. Mutants which reduced the kinase:phosphatase ratio had the opposite effect. These results indicate that the main effect of the bifunctional enzyme on islet pulsatility is due to Fru2,6-BP alteration of the threshold for autocatalytic activation of PFK1 by Fru1,6-BP. Using computational models based on PFK1-generated islet oscillations, we then illustrated how moderate elevation of Fru-2,6-BP can increase the frequency of glycolytic oscillations while reducing their amplitude, with sufficiently high activation resulting in termination of slow oscillations. The concordance we observed between PFK2/FBPase2-induced modulation of islet oscillations and the models of PFK1-driven oscillations furthermore suggests that metabolic oscillations, like those found in yeast and skeletal muscle, are shaped early in glycolysis.


Biophysical Journal | 2016

Phase Analysis of Metabolic Oscillations and Membrane Potential in Pancreatic Islet β-Cells

Matthew J. Merrins; Chetan Poudel; Joseph P. McKenna; Joon Ha; Arthur Sherman; Richard Bertram; Leslie S. Satin

Metabolism in islet β-cells displays oscillations that can trigger pulses of electrical activity and insulin secretion. There has been a decades-long debate among islet biologists about whether metabolic oscillations are intrinsic or occur in response to oscillations in intracellular Ca(2+) that result from bursting electrical activity. In this article, the dynamics of oscillatory metabolism were investigated using five different optical reporters. Reporter activity was measured simultaneously with membrane potential bursting to determine the phase relationships between the metabolic oscillations and electrical activity. Our experimental findings suggest that Ca(2+) entry into β-cells stimulates the rate of mitochondrial metabolism, accounting for the depletion of glycolytic intermediates during each oscillatory burst. We also performed Ca(2+) clamp tests in which we clamped membrane potential with the KATP channel-opener diazoxide and KCl to fix Ca(2+) at an elevated level. These tests confirm that metabolic oscillations do not require Ca(2+) oscillations, but show that Ca(2+) plays a larger role in shaping metabolic oscillations than previously suspected. A dynamical picture of the mechanisms of oscillations emerged that requires the restructuring of contemporary mathematical β-cell models, including our own dual oscillator model. In the companion article, we modified our model to account for these new data.


Journal of Biological Chemistry | 2014

Pancreatic and Duodenal Homeobox Protein 1 (Pdx-1) Maintains Endoplasmic Reticulum Calcium Levels through Transcriptional Regulation of Sarco-endoplasmic Reticulum Calcium ATPase 2b (SERCA2b) in the Islet β Cell

Justin S. Johnson; Tatsuyoshi Kono; Xin Tong; Wataru Yamamoto; Angel Zarain-Herzberg; Matthew J. Merrins; Leslie S. Satin; Patrick Gilon; Carmella Evans-Molina

Background: Altered sarco-endoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) expression and activity contributes to β cell dysfunction in diabetes. Results: SERCA2b deficiency occurs secondary to loss of pancreatic and duodenal homeobox 1 (Pdx-1)-mediated transcriptional regulation. Conclusion: Pdx-1 maintains SERCA2b expression and endoplasmic reticulum (ER) calcium levels in the β cell. Significance: These findings elucidate a novel pathway that contributes to β cell ER stress. Although the pancreatic duodenal homeobox 1 (Pdx-1) transcription factor is known to play an indispensable role in β cell development and secretory function, recent data also implicate Pdx-1 in the maintenance of endoplasmic reticulum (ER) health. The sarco-endoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) pump maintains a steep Ca2+ gradient between the cytosol and ER lumen. In models of diabetes, our data demonstrated loss of β cell Pdx-1 that occurs in parallel with altered SERCA2b expression, whereas in silico analysis of the SERCA2b promoter revealed multiple putative Pdx-1 binding sites. We hypothesized that Pdx-1 loss under inflammatory and diabetic conditions leads to decreased SERCA2b levels and activity with concomitant alterations in ER health. To test this, siRNA-mediated knockdown of Pdx-1 was performed in INS-1 cells. The results revealed reduced SERCA2b expression and decreased ER Ca2+, which was measured using fluorescence lifetime imaging microscopy. Cotransfection of human Pdx-1 with a reporter fused to the human SERCA2 promoter increased luciferase activity 3- to 4-fold relative to an empty vector control, and direct binding of Pdx-1 to the proximal SERCA2 promoter was confirmed by chromatin immunoprecipitation. To determine whether restoration of SERCA2b could rescue ER stress induced by Pdx-1 loss, Pdx1+/− mice were fed a high-fat diet. Isolated islets demonstrated an increased spliced-to-total Xbp1 ratio, whereas SERCA2b overexpression reduced the Xbp1 ratio to that of wild-type controls. Together, these results identify SERCA2b as a novel transcriptional target of Pdx-1 and define a role for altered ER Ca2+ regulation in Pdx-1-deficient states.


The Journal of Physiology | 2018

Restoration of metabolic health by decreased consumption of branched‐chain amino acids

Nicole E. Cummings; Elizabeth M. Williams; Ildiko Kasza; Elizabeth N. Konon; Michael D. Schaid; Brian A. Schmidt; Chetan Poudel; Dawn S. Sherman; Deyang Yu; Sebastian I. Arriola Apelo; Sara E. Cottrell; Gabriella Geiger; Macy E. Barnes; Jaclyn A. Wisinski; Rachel J. Fenske; Kristina A. Matkowskyj; Michelle E. Kimple; Caroline M. Alexander; Matthew J. Merrins; Dudley W. Lamming

We recently found that feeding healthy mice a diet with reduced levels of branched-chain amino acids (BCAAs), which are associated with insulin resistance in both humans and rodents, modestly improves glucose tolerance and slows fat mass gain. In the present study, we show that a reduced BCAA diet promotes rapid fat mass loss without calorie restriction in obese mice. Selective reduction of dietary BCAAs also restores glucose tolerance and insulin sensitivity to obese mice, even as they continue to consume a high‐fat, high‐sugar diet. A low BCAA diet transiently induces FGF21 (fibroblast growth factor 21) and increases energy expenditure. We suggest that dietary protein quality (i.e. the precise macronutrient composition of dietary protein) may impact the effectiveness of weight loss diets.


Diabetes | 2016

Pancreatic β cells from Mice Offset Age-Associated Mitochondrial Deficiency with Reduced KATP Channel Activity

Trillian Gregg; Chetan Poudel; Brian A. Schmidt; Rashpal S. Dhillon; Sophia M. Sdao; Nathan A. Truchan; Emma L. Baar; Luis A. Fernandez; John M. Denu; Kevin W. Eliceiri; Jeremy D. Rogers; Michelle E. Kimple; Dudley W. Lamming; Matthew J. Merrins

Aging is accompanied by impaired glucose homeostasis and an increased risk of type 2 diabetes, culminating in the failure of insulin secretion from pancreatic β-cells. To investigate the effects of age on β-cell metabolism, we established a novel assay to directly image islet metabolism with NAD(P)H fluorescence lifetime imaging (FLIM). We determined that impaired mitochondrial activity underlies an age-dependent loss of insulin secretion in human islets. NAD(P)H FLIM revealed a comparable decline in mitochondrial function in the pancreatic islets of aged mice (≥24 months), the result of 52% and 57% defects in flux through complex I and II, respectively, of the electron transport chain. However, insulin secretion and glucose tolerance are preserved in aged mouse islets by the heightened metabolic sensitivity of the β-cell triggering pathway, an adaptation clearly encoded in the metabolic and Ca2+ oscillations that trigger insulin release (Ca2+ plateau fraction: young 0.211 ± 0.006, aged 0.380 ± 0.007, P < 0.0001). This enhanced sensitivity is driven by a reduction in KATP channel conductance (diazoxide: young 5.1 ± 0.2 nS; aged 3.5 ± 0.5 nS, P < 0.01), resulting in an ∼2.8 mmol/L left shift in the β-cell glucose threshold. The results demonstrate how mice but not humans are able to successfully compensate for age-associated metabolic dysfunction by adjusting β-cell glucose sensitivity and highlight an essential mechanism for ensuring the maintenance of insulin secretion.

Collaboration


Dive into the Matthew J. Merrins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arthur Sherman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brian A. Schmidt

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Chetan Poudel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michelle E. Kimple

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dudley W. Lamming

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Nicole E. Cummings

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rachel J. Fenske

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Caroline M. Alexander

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge