Network


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

Hotspot


Dive into the research topics where Heather W. Collins is active.

Publication


Featured researches published by Heather W. Collins.


Journal of Biological Chemistry | 2003

Regulation of Leucine-stimulated Insulin Secretion and Glutamine Metabolism in Isolated Rat Islets

Changhong Li; Habiba Najafi; Yevgeny Daikhin; Ilana Nissim; Heather W. Collins; Marc Yudkoff; Franz M. Matschinsky; Charles A. Stanley

Glutamate dehydrogenase (GDH) is regulated by both positive (leucine and ADP) and negative (GTP and ATP) allosteric factors. We hypothesized that the phosphate potential of β-cells regulates the sensitivity of leucine stimulation. These predictions were tested by measuring leucine-stimulated insulin secretion in perifused rat islets following glucose depletion and by tracing the nitrogen flux of [2-15N]glutamine using stable isotope techniques. The sensitivity of leucine stimulation was enhanced by long time (120-min) energy depletion and inhibited by glucose pretreatment. After limited 50-min glucose depletion, leucine, not α-ketoisocaproate, failed to stimulate insulin release. β-Cells sensitivity to leucine is therefore proposed to be a function of GDH activation. Leucine increased the flux through GDH 3-fold compared with controls while causing insulin release. High glucose inhibited flux through both glutaminase and GDH, and leucine was unable to override this inhibition. These results clearly show that leucine induced the secretion of insulin by augmenting glutaminolysis through activating glutaminase and GDH. Glucose regulates β-cell sensitivity to leucine by elevating the ratio of ATP and GTP to ADP and Pi and thereby decreasing the flux through GDH and glutaminase. These mechanisms provide an explanation for hypoglycemia caused by mutations of GDH in children.


Journal of Biological Chemistry | 2006

Green Tea Polyphenols Modulate Insulin Secretion by Inhibiting Glutamate Dehydrogenase

Changhong Li; Aron Allen; Jae Kwagh; Nicolai M. Doliba; Wei Qin; Habiba Najafi; Heather W. Collins; Franz M. Matschinsky; Charles A. Stanley; Thomas J. Smith

Insulin secretion by pancreatic β-cells is stimulated by glucose, amino acids, and other metabolic fuels. Glutamate dehydrogenase (GDH) has been shown to play a regulatory role in this process. The importance of GDH was underscored by features of hyperinsulinemia/hyperammonemia syndrome, where a dominant mutation causes the loss of inhibition by GTP and ATP. Here we report the effects of green tea polyphenols on GDH and insulin secretion. Of the four compounds tested, epigallocatechin gallate (EGCG) and epicatechin gallate were found to inhibit GDH with nanomolar ED50 values and were therefore found to be as potent as the physiologically important inhibitor GTP. Furthermore, we have demonstrated that EGCG inhibits BCH-stimulated insulin secretion, a process that is mediated by GDH, under conditions where GDH is no longer inhibited by high energy metabolites. EGCG does not affect glucose-stimulated insulin secretion under high energy conditions where GDH is probably fully inhibited. We have further shown that these compounds act in an allosteric manner independent of their antioxidant activity and that the β-cell stimulatory effects are directly correlated with glutamine oxidation. These results demonstrate that EGCG, much like the activator of GDH (BCH), can facilitate dissecting the complex regulation of insulin secretion by pharmacologically modulating the effects of GDH.


Journal of Biological Chemistry | 2010

Mechanism of Hyperinsulinism in Short-chain 3-Hydroxyacyl-CoA Dehydrogenase Deficiency Involves Activation of Glutamate Dehydrogenase

Changhong Li; Pan Chen; Andrew A. Palladino; Srinivas B. Narayan; Laurie K. Russell; Samir Sayed; Guoxiang Xiong; Jie Chen; David Stokes; Yasmeen Butt; Patricia M. Jones; Heather W. Collins; Noam A. Cohen; Akiva S. Cohen; Itzhak Nissim; Thomas J. Smith; Arnold W. Strauss; Franz M. Matschinsky; Michael Bennett; Charles A. Stanley

The mechanism of insulin dysregulation in children with hyperinsulinism associated with inactivating mutations of short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD) was examined in mice with a knock-out of the hadh gene (hadh−/−). The hadh−/− mice had reduced levels of plasma glucose and elevated plasma insulin levels, similar to children with SCHAD deficiency. hadh−/− mice were hypersensitive to oral amino acid with decrease of glucose level and elevation of insulin. Hypersensitivity to oral amino acid in hadh−/− mice can be explained by abnormal insulin responses to a physiological mixture of amino acids and increased sensitivity to leucine stimulation in isolated perifused islets. Measurement of cytosolic calcium showed normal basal levels and abnormal responses to amino acids in hadh−/− islets. Leucine, glutamine, and alanine are responsible for amino acid hypersensitivity in islets. hadh−/− islets have lower intracellular glutamate and aspartate levels, and this decrease can be prevented by high glucose. hadh−/− islets also have increased [U-14C]glutamine oxidation. In contrast, hadh−/− mice have similar glucose tolerance and insulin sensitivity compared with controls. Perifused hadh−/− islets showed no differences from controls in response to glucose-stimulated insulin secretion, even with addition of either a medium-chain fatty acid (octanoate) or a long-chain fatty acid (palmitate). Pull-down experiments with SCHAD, anti-SCHAD, or anti-GDH antibodies showed protein-protein interactions between SCHAD and GDH. GDH enzyme kinetics of hadh−/− islets showed an increase in GDH affinity for its substrate, α-ketoglutarate. These studies indicate that SCHAD deficiency causes hyperinsulinism by activation of GDH via loss of inhibitory regulation of GDH by SCHAD.


Journal of Biological Chemistry | 2009

Transforming Growth Factor-β/Smad3 Signaling Regulates Insulin Gene Transcription and Pancreatic Islet β-Cell Function

Huei-Min Lin; Ji-Hyeon Lee; Hariom Yadav; Anil K. Kamaraju; Eric Liu; Duan Zhigang; Anthony Vieira; Seong-Jin Kim; Heather W. Collins; Franz M. Matschinsky; David M. Harlan; Anita B. Roberts; Sushil G. Rane

Pancreatic islet β-cell dysfunction is a signature feature of Type 2 diabetes pathogenesis. Consequently, knowledge of signals that regulate β-cell function is of immense clinical relevance. Transforming growth factor (TGF)-β signaling plays a critical role in pancreatic development although the role of this pathway in the adult pancreas is obscure. Here, we define an important role of the TGF-β pathway in regulation of insulin gene transcription and β-cell function. We identify insulin as a TGF-β target gene and show that the TGF-β signaling effector Smad3 occupies the insulin gene promoter and represses insulin gene transcription. In contrast, Smad3 small interfering RNAs relieve insulin transcriptional repression and enhance insulin levels. Transduction of adenoviral Smad3 into primary human and non-human primate islets suppresses insulin content, whereas, dominant-negative Smad3 enhances insulin levels. Consistent with this, Smad3-deficient mice exhibit moderate hyperinsulinemia and mild hypoglycemia. Moreover, Smad3 deficiency results in improved glucose tolerance and enhanced glucose-stimulated insulin secretion in vivo. In ex vivo perifusion assays, Smad3-deficient islets exhibit improved glucose-stimulated insulin release. Interestingly, Smad3-deficient islets harbor an activated insulin-receptor signaling pathway and TGF-β signaling regulates expression of genes involved in β-cell function. Together, these studies emphasize TGF-β/Smad3 signaling as an important regulator of insulin gene transcription and β-cell function and suggest that components of the TGF-β signaling pathway may be dysregulated in diabetes.


Journal of Biological Chemistry | 2006

Effects of a GTP-insensitive Mutation of Glutamate Dehydrogenase on Insulin Secretion in Transgenic Mice

Changhong Li; Andrea Matter; Andrea Kelly; Tom J. Petty; Habiba Najafi; Courtney MacMullen; Yevgeny Daikhin; Ilana Nissim; Adam Lazarow; Jae Kwagh; Heather W. Collins; Betty Y.L. Hsu; Itzhak Nissim; Marc Yudkoff; Franz M. Matschinsky; Charles A. Stanley

Glutamate dehydrogenase (GDH) plays an important role in insulin secretion as evidenced in children by gain of function mutations of this enzyme that cause a hyperinsulinism-hyperammonemia syndrome (GDH-HI) and sensitize β-cells to leucine stimulation. GDH transgenic mice were generated to express the human GDH-HI H454Y mutation and human wild-type GDH in islets driven by the rat insulin promoter. H454Y transgene expression was confirmed by increased GDH enzyme activity in islets and decreased sensitivity to GTP inhibition. The H454Y GDH transgenic mice had hypoglycemia with normal growth rates. H454Y GDH transgenic islets were more sensitive to leucine- and glutamine-stimulated insulin secretion but had decreased response to glucose stimulation. The fluxes via GDH and glutaminase were measured by tracing 15N flux from [2-15N]glutamine. The H454Y transgene in islets had higher insulin secretion in response to glutamine alone and had 2-fold greater GDH flux. High glucose inhibited both glutaminase and GDH flux, and leucine could not override this inhibition. 15NH4Cl tracing studies showed 15N was not incorporated into glutamate in either H454Y transgenic or normal islets. In conclusion, we generated a GDH-HI disease mouse model that has a hypoglycemia phenotype and confirmed that the mutation of H454Y is disease causing. Stimulation of insulin release by the H454Y GDH mutation or by leucine activation is associated with increased oxidative deamination of glutamate via GDH. This study suggests that GDH functions predominantly in the direction of glutamate oxidation rather than glutamate synthesis in mouse islets and that this flux is tightly controlled by glucose.


Diabetes | 1996

Wortmannin Inhibits Insulin Secretion in Pancreatic Islets and β-TC3 Cells Independent of Its Inhibition of Phosphatidylinositol 3-Kinase

Zhiyong Gao; Robert J. Konrad; Heather W. Collins; Franz M. Matschinsky; Paul Rothenberg; Bryan A. Wolf

Glucose is the primary stimulus for insulin secretion by pancreatic β-cells, and it triggers membrane depolarization and influx of extracellular Ca2+. Cholinergic agonists amplify insulin release by several pathways, including activation of phospholipase C, which hydrolyzes membrane polyphosphoinositides. A novel phospholipid, phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3[, a product of phosphatidylinositol 3-kinase (PI 3-kinase), has recently been found in various cell types. We demonstrate by immunoblotting that PI 3-kinase is present in both cytosolic and membrane fractions of insulin-secreting β-TC3 cells and in rat islets. The catalytic activity of PI 3-kinase in immunoprecipitates of islets and β-TC3 cells was measured by the production of radioactive phosphatidylinositol 3-monophosphate from phosphatidylinositol (Ptdlns) in the presence of [γ-32P[ATP. Wortmannin, a fungal metabolite, dose dependency inhibited PI 3-kinase activity of both islets and P-TC3 cells, with an IC50 of 1 nmol/l and a maximally effective concentration of 100 nmol/l, when it was added directly to the kinase assay. However, if intact islets were incubated with wortmannin and PI 3-kinase subsequently was determined in islet immunoprecipitates, ∼50% inhibition of PI 3-kinase activity (but no inhibition of glucose- and carbachol-stimulated insulin secretion) from intact islets was obtained at wortmannin concentrations of 100 µmol/l. Wortmannin, at higher concentrations (1 and 10 µmol/l), inhibited glucose- and carbachol-induced insulin secretion of intact rat islets by 58 and 92%, respectively. Wortmannin had no effect on the basal insulin release from rat islets. A similar dose curve of inhibition of glucose- and carbachol-induced insulin secretion by wortmannin was obtained when β-TC3 cells were used. Cellu-lar metabolism was not changed by any wortmannin concentrations tested (0.01–10 µmol/l). Both basal cytosolic [Ca2+]1 and carbamyl choline-induced increases of [Ca2+]1 were unaffected by wortmannin in the presence of 2.5 mmol/l Ca2+, while Ca2+ mobilization from intracellular stores was partially decreased by wortmannin. Together, these data suggest that wortmannin at concentrations that inhibit PI 3-kinase does not affect insulin secretion. PI 3-kinase is unlikely to have a major role in insulin secretion induced by glucose and carbachol.Diabetes 45:854–862, 1996


Journal of Biological Chemistry | 2008

Elimination of KATP Channels in Mouse Islets Results in Elevated [U-13C]Glucose Metabolism, Glutaminolysis, and Pyruvate Cycling but a Decreased γ-Aminobutyric Acid Shunt

Changhong Li; Itzhak Nissim; Pan Chen; Carol Buettger; Habiba Najafi; Yevgeny Daikhin; Ilana Nissim; Heather W. Collins; Marc Yudkoff; Charles A. Stanley; Franz M. Matschinsky

Pancreatic beta cells are hyper-responsive to amino acids but have decreased glucose sensitivity after deletion of the sulfonylurea receptor 1 (SUR1) both in man and mouse. It was hypothesized that these defects are the consequence of impaired integration of amino acid, glucose, and energy metabolism in beta cells. We used gas chromatography-mass spectrometry methodology to study intermediary metabolism of SUR1 knock-out (SUR1-/-) and control mouse islets with d-[U-13C]glucose as substrate and related the results to insulin secretion. The levels and isotope labeling of alanine, aspartate, glutamate, glutamine, and γ-aminobutyric acid (GABA) served as indicators of intermediary metabolism. We found that the GABA shunt of SUR1-/- islets is blocked by about 75% and showed that this defect is due to decreased glutamate decarboxylase synthesis, probably caused by elevated free intracellular calcium. Glutaminolysis stimulated by the leucine analogue d,l-β-2-amino-2-norbornane-carboxylic acid was, however, enhanced in SUR1-/- and glyburide-treated SUR1+/+ islets. Glucose oxidation and pyruvate cycling was increased in SUR1-/- islets at low glucose but was the same as in controls at high glucose. Malic enzyme isoforms 1, 2, and 3, involved in pyruvate cycling, were all expressed in islets. High glucose lowered aspartate and stimulated glutamine synthesis similarly in controls and SUR1-/- islets. The data suggest that the interruption of the GABA shunt and the lack of glucose regulation of pyruvate cycling may cause the glucose insensitivity of the SUR1-/- islets but that enhanced basal pyruvate cycling, lowered GABA shunt flux, and enhanced glutaminolytic capacity may sensitize the beta cells to amino acid stimulation.


Journal of Biological Chemistry | 2007

Glucokinase Thermolability and Hepatic Regulatory Protein Binding Are Essential Factors for Predicting the Blood Glucose Phenotype of Missense Mutations

Maria Pino; Kyoung-Ah Kim; Kathy D. Shelton; Jill Lindner; Stella Odili; Changhong Li; Heather W. Collins; Masakazu Shiota; Franz M. Matschinsky; Mark A. Magnuson

To better understand how glucokinase (GK) missense mutations associated with human glycemic diseases perturb glucose homeostasis, we generated and characterized mice with either an activating (A456V) or inactivating (K414E) mutation in the gk gene. Animals with these mutations exhibited alterations in their blood glucose concentration that were inversely related to the relative activity index of GK. Moreover, the threshold for glucose-stimulated insulin secretion from islets with either the activating or inactivating mutation were left- or right-shifted, respectively. However, we were surprised to find that mice with the activating mutation had markedly reduced amounts of hepatic GK activity. Further studies of bacterially expressed mutant enzymes revealed that GKA456V is as stable as the wild type enzyme, whereas GKK414E is thermolabile. However, the ability of GK regulatory protein to inhibit GKA456V was found to be less than that of the wild type enzyme, a finding consistent with impaired hepatic nuclear localization. Taken together, this study indicates that it is necessary to have knowledge of both thermolability and the interactions of mutant GK enzymes with GK regulatory protein when attempting to predict in vivo glycemic phenotypes based on the measurement of enzyme kinetics.


European Journal of Neuroscience | 1998

Mechanisms of spontaneous cytosolic Ca2+ transients in differentiated human neuronal cells

Zhiyong Gao; Meng Chen; Heather W. Collins; Franz M. Matschinsky; Virginia M.-Y. Lee; Bryan A. Wolf

We have studied Ca2+ homeostasis in a unique model of human neurons, the NT2N cell, which differentiates from a human teratocarcinoma cell line, NTera2/C1.D1 by retinoic acid treatment. When perifused with Krebs–HEPES buffer containing 2.5 mm CaCl2, fura‐2 loaded NT2N cells produced spontaneous cytosolic Ca2+ oscillations, or Ca2+ transients. These cytosolic Ca2+ transients were not blocked by antagonists of glutamate (6‐cyano‐7‐nitroquinoxaline‐2,3‐dione and d(–)‐2‐amino‐5‐phosphonopentanoic acid) or muscarinic (atropine) receptors. Omission of extracellular Ca2+ completely abolished Ca2+ oscillations and decreased the average Ca2+ level from 106 ± 14 nm to 59 ± 8 nm. Addition of the L‐type Ca2+ channel blocker nifedipine (1 or 10 μm) or of the N‐type inhibitor ω‐conotoxin GVIA (5 μm) significantly, although incompletely, suppressed Ca2+ oscillations, while ω‐conotoxin MVIIC (5 μm), a selective antagonist of P‐ and Q‐channels, had no effect. Ni2+, at 100 μm, a concentration selective for T‐type channels, did not inhibit Ca2+ transients. Non‐specific blockage of Ca2+ channels by higher concentrations of Ni2+ (2–5 mm) or Co2+ (1 mm) abolished Ca2+ oscillations completely. The endoplasmic reticulum Ca2+‐ATPase inhibitor, thapsigargin (1 μm), slightly decreased Ca2+ oscillation frequency, and induced a small transitory increase in the average cytosolic Ca2+ concentration. The mRNAs of L‐ (α1D subunit) and N‐type (α1B subunit) Ca2+ channel were present in NT2N cells, while that of a T‐type Ca2+ channel (α1‐subunit) was not present in the NT2N cells as shown by reverse transcription–polymerase chain reaction. In conclusion, NT2N neuronal cells generate cytosolic Ca2+ oscillations mainly by influx of extracellular Ca2+ through multiple channels, which include L‐ and N‐type channels, and do not require activation of glutamate or muscarinic receptors.


The New England Journal of Medicine | 2008

Amyloid and transplanted islets.

Michael R. Rickels; Heather W. Collins; Ali Naji

TO THE EDITOR: In their letter to the editor, Westermark and colleagues (Aug. 28 issue)1 report on their identification of amyloid in 43% of intrahepatically transplanted islets on postmortem examination of a recipient with type 1 diabetes. Amyloid is composed of amylin (islet amyloid polypeptide [IAPP]) that is cosecreted from the beta cell with insulin but normally is inhibited from forming amyloid by appropriate proportions of insulin and other factors in the beta cell.2,3 Proportions of insulin and amylin within the beta cell are best estimated in vivo following secretion after acute stimulation.4 To determine whether insulin and amylin are secreted in appropriate proportions after human islet transplantation, we measured plasma concentrations of both hormones before and after arginine stimulation under fasting and hyperglycemic-clamp conditions in four insulin-independent islet recipients and matched controls; the characteristics and C-peptide data of the subjects were reported previously.5 Insulin responses to arginine were relatively more reduced during the hyperglycemic clamps than were amylin responses, resulting in markedly lower ratios between insulin and amylin (Fig. 1). These data indicate that during hyperglycemia, intrahepatically transplanted islets secrete disproportionately more amylin than normal, suggesting that hyperglycemia in the islet recipient may have contributed to the observed amyloid deposition. Figure 1 Plasma Insulin, Amylin, and Molar Ratios between Insulin and Amylin in Four Insulin-Independent Recipients of Islet Transplantation and Four Control Subjects without Diabetes

Collaboration


Dive into the Heather W. Collins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carol Buettger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Changhong Li

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Charles A. Stanley

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Habiba Najafi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Nicolai M. Doliba

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wei Qin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ilana Nissim

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Marc Yudkoff

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge