167 results on '"Jesper Gromada"'
Search Results
2. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance
- Author
-
Rene C. Adam, Ivory J. Mintah, Corey A. Alexa-Braun, Lisa M. Shihanian, Joseph S. Lee, Poulabi Banerjee, Sara C. Hamon, Hye In Kim, Jonathan C. Cohen, Helen H. Hobbs, Cristopher Van Hout, Jesper Gromada, Andrew J. Murphy, George D. Yancopoulos, Mark W. Sleeman, and Viktoria Gusarova
- Subjects
atherosclerosis ,cardiovascular disease ,familial hypercholesterolemia ,low density lipoprotein-cholesterol ,low density lipoprotein receptor ,lipidomics ,Biochemistry ,QD415-436 - Abstract
Angiopoietin-like protein (ANGPTL)3 regulates plasma lipids by inhibiting LPL and endothelial lipase (EL). ANGPTL3 inactivation lowers LDL-C independently of the classical LDLR-mediated pathway and represents a promising therapeutic approach for individuals with homozygous familial hypercholesterolemia due to LDLR mutations. Yet, how ANGPTL3 regulates LDL-C levels is unknown. Here, we demonstrate in hyperlipidemic humans and mice that ANGPTL3 controls VLDL catabolism upstream of LDL. Using kinetic, lipidomic, and biophysical studies, we show that ANGPTL3 inhibition reduces VLDL-lipid content and size, generating remnant particles that are efficiently removed from the circulation. This suggests that ANGPTL3 inhibition lowers LDL-C by limiting LDL particle production. Mechanistically, we discovered that EL is a key mediator of ANGPTL3's novel pathway. Our experiments revealed that, although dispensable in the presence of LDLR, EL-mediated processing of VLDL becomes critical for LDLR-independent particle clearance. In the absence of EL and LDLR, ANGPTL3 inhibition perturbed VLDL catabolism, promoted accumulation of atypical remnants, and failed to reduce LDL-C. Taken together, we uncover ANGPTL3 at the helm of a novel EL-dependent pathway that lowers LDL-C in the absence of LDLR.
- Published
- 2020
- Full Text
- View/download PDF
3. Alirocumab, evinacumab, and atorvastatin triple therapy regresses plaque lesions and improves lesion composition in mice[S]
- Author
-
Marianne G. Pouwer, Elsbet J. Pieterman, Nicole Worms, Nanda Keijzer, J. Wouter Jukema, Jesper Gromada, Viktoria Gusarova, and Hans M.G. Princen
- Subjects
atherosclerosis ,drug therapy/hypolipidemic drugs ,macrophages/monocytes ,apolipoproteins ,antibodies ,regression ,Biochemistry ,QD415-436 - Abstract
Atherosclerosis-related CVD causes nearly 20 million deaths annually. Most patients are treated after plaques develop, so therapies must regress existing lesions. Current therapies reduce plaque volume, but targeting all apoB-containing lipoproteins with intensive combinations that include alirocumab or evinacumab, monoclonal antibodies against cholesterol-regulating proprotein convertase subtilisin/kexin type 9 and angiopoietin-like protein 3, may provide more benefit. We investigated the effect of such lipid-lowering interventions on atherosclerosis in APOE*3-Leiden.CETP mice, a well-established model for hyperlipidemia. Mice were fed a Western-type diet for 13 weeks and thereafter matched into a baseline group (euthanized at 13 weeks) and five groups that received diet alone (control) or with treatment [atorvastatin; atorvastatin and alirocumab; atorvastatin and evinacumab; or atorvastatin, alirocumab, and evinacumab (triple therapy)] for 25 weeks. We measured effects on cholesterol levels, plaque composition and morphology, monocyte adherence, and macrophage proliferation. All interventions reduced plasma total cholesterol (37% with atorvastatin to 80% with triple treatment; all P < 0.001). Triple treatment decreased non-HDL-C to 1.0 mmol/l (91% difference from control; P < 0.001). Atorvastatin reduced atherosclerosis progression by 28% versus control (P < 0.001); double treatment completely blocked progression and diminished lesion severity. Triple treatment regressed lesion size versus baseline in the thoracic aorta by 50% and the aortic root by 36% (both P < 0.05 vs. baseline), decreased macrophage accumulation through reduced proliferation, and abated lesion severity. Thus, high-intensive cholesterol-lowering triple treatment targeting all apoB-containing lipoproteins regresses atherosclerotic lesion area and improves lesion composition in mice, making it a promising potential approach for treating atherosclerosis.
- Published
- 2020
- Full Text
- View/download PDF
4. RNA-sequencing reveals altered skeletal muscle contraction, E3 ligases, autophagy, apoptosis, and chaperone expression in patients with critical illness myopathy
- Author
-
Monica Llano-Diez, Wen Fury, Haruka Okamoto, Yu Bai, Jesper Gromada, and Lars Larsson
- Subjects
Critical illness myopathy ,RNA sequencing ,Skeletal muscle transcriptomics ,Gene expression ,Intensive care unit ,Mechanical loading ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Critical illness myopathy (CIM) is associated with severe skeletal muscle wasting and impaired function in intensive care unit (ICU) patients. The mechanisms underlying CIM remain incompletely understood. To elucidate the biological activities occurring at the transcriptional level in the skeletal muscle of ICU patients with CIM, the gene expression profiles, potential upstream regulators, and enrichment pathways were characterized using RNA sequencing (RNA-seq). We also compared the skeletal muscle gene signatures in ICU patients with CIM and genes perturbed by mechanical loading in one leg of the ICU patients, with an aim of reducing the loss of muscle function. Methods RNA-seq was used to assess gene expression changes in tibialis anterior skeletal muscle samples from seven critically ill, immobilized, and mechanically ventilated ICU patients with CIM and matched control subjects. We also examined skeletal muscle gene expression for both legs of six ICU patients with CIM, where one leg was mechanically loaded for 10 h/day for an average of 9 days. Results In total, 6257 of 17,221 detected genes were differentially expressed (84% upregulated; p
- Published
- 2019
- Full Text
- View/download PDF
5. Genetic inactivation of ANGPTL4 improves glucose homeostasis and is associated with reduced risk of diabetes
- Author
-
Viktoria Gusarova, Colm O’Dushlaine, Tanya M. Teslovich, Peter N. Benotti, Tooraj Mirshahi, Omri Gottesman, Cristopher V. Van Hout, Michael F. Murray, Anubha Mahajan, Jonas B. Nielsen, Lars Fritsche, Anders Berg Wulff, Daniel F. Gudbjartsson, Marketa Sjögren, Connor A. Emdin, Robert A. Scott, Wen-Jane Lee, Aeron Small, Lydia C. Kwee, Om Prakash Dwivedi, Rashmi B. Prasad, Shannon Bruse, Alexander E. Lopez, John Penn, Anthony Marcketta, Joseph B. Leader, Christopher D. Still, H. Lester Kirchner, Uyenlinh L. Mirshahi, Amr H. Wardeh, Cassandra M. Hartle, Lukas Habegger, Samantha N. Fetterolf, Teresa Tusie-Luna, Andrew P. Morris, Hilma Holm, Valgerdur Steinthorsdottir, Patrick Sulem, Unnur Thorsteinsdottir, Jerome I. Rotter, Lee-Ming Chuang, Scott Damrauer, David Birtwell, Chad M. Brummett, Amit V. Khera, Pradeep Natarajan, Marju Orho-Melander, Jason Flannick, Luca A. Lotta, Cristen J. Willer, Oddgeir L. Holmen, Marylyn D. Ritchie, David H. Ledbetter, Andrew J. Murphy, Ingrid B. Borecki, Jeffrey G. Reid, John D. Overton, Ola Hansson, Leif Groop, Svati H. Shah, William E. Kraus, Daniel J. Rader, Yii-Der I. Chen, Kristian Hveem, Nicholas J. Wareham, Sekar Kathiresan, Olle Melander, Kari Stefansson, Børge G. Nordestgaard, Anne Tybjærg-Hansen, Goncalo R. Abecasis, David Altshuler, Jose C. Florez, Michael Boehnke, Mark I. McCarthy, George D. Yancopoulos, David J. Carey, Alan R. Shuldiner, Aris Baras, Frederick E. Dewey, and Jesper Gromada
- Subjects
Science - Abstract
Genetic variation in ANGPTL4 is associated with lipid traits. Here, the authors find that predicted loss-of-function variants in ANGPTL4 are associated with glucose homeostasis and reduced risk of type 2 diabetes and that Angptl4 −/− mice on a high-fat diet show improved insulin sensitivity.
- Published
- 2018
- Full Text
- View/download PDF
6. Inhibition of PCSK9 does not improve lipopolysaccharide-induced mortality in mice
- Author
-
Jean-Mathieu Berger, Angel Loza Valdes, Jesper Gromada, Norma Anderson, and Jay D. Horton
- Subjects
lipopolysaccharide ,endotoxemia ,cholesterol ,LDL ,PCSK9 ,Biochemistry ,QD415-436 - Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that targets LDL receptors (LDLRs) for degradation in liver. Blocking the interaction of PCSK9 with the LDLR potently reduces plasma LDL cholesterol levels and cardiovascular events. Recently, it has been suggested that inhibition of PCSK9 might also improve outcomes in mice and humans with sepsis, possibly by increasing LDLR-mediated clearance of endotoxins. Sepsis is a complication of a severe microbial infection that has shared pathways with lipid metabolism. Here, we tested whether anti-PCSK9 antibodies prevent death from lipopolysaccharide (LPS)-induced endotoxemia. Mice were administered PCSK9 antibodies prior to, or shortly after, injecting LPS. In both scenarios, the administration of PCSK9 antibodies did not alter endotoxemia-induced mortality. Afterward, we determined whether the complete absence of PCSK9 improved endotoxemia-induced mortality in mice with the germ-line deletion of Pcsk9. Similarly, PCSK9 knockout mice were not protected from LPS-induced death. To determine whether low LDLR expression increased LPS-induced mortality, Ldlr−/− mice and PCSK9 transgenic mice were studied after injection of LPS. Endotoxemia-induced mortality was not altered in either mouse model. In a human cohort, we observed no correlation between plasma inflammation markers with total cholesterol levels, LDL cholesterol, and PCSK9. Combined, our data demonstrate that PCSK9 inhibition provides no protection from LPS-induced mortality in mice.
- Published
- 2017
- Full Text
- View/download PDF
7. ANGPTL8 requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clearance[S]
- Author
-
Jorge F. Haller, Ivory J. Mintah, Lisa M. Shihanian, Panayiotis Stevis, David Buckler, Corey A. Alexa-Braun, Sandra Kleiner, Serena Banfi, Jonathan C. Cohen, Helen H. Hobbs, George D. Yancopoulos, Andrew J. Murphy, Viktoria Gusarova, and Jesper Gromada
- Subjects
angiopoietin-like 8 ,angiopoietin-like 3 ,blocking antibody ,Biochemistry ,QD415-436 - Abstract
Angiopoietin-like (ANGPTL)3 and ANGPTL8 are secreted proteins and inhibitors of LPL-mediated plasma triglyceride (TG) clearance. It is unclear how these two ANGPTL proteins interact to regulate LPL activity. ANGPTL3 inhibits LPL activity and increases serum TG independent of ANGPTL8. These effects are reversed with an ANGPTL3 blocking antibody. Here, we show that ANGPTL8, although it possesses a functional inhibitory motif, is inactive by itself and requires ANGPTL3 expression to inhibit LPL and increase plasma TG. Using a mutated form of ANGPTL3 that lacks LPL inhibitory activity, we demonstrate that ANGPTL3 activity is not required for its ability to activate ANGPTL8. Moreover, coexpression of ANGPTL3 and ANGPTL8 leads to a far more efficacious increase in TG in mice than ANGPTL3 alone, suggesting the major inhibitory activity of this complex derives from ANGPTL8. An antibody to the C terminus of ANGPTL8 reversed LPL inhibition by ANGPTL8 in the presence of ANGPTL3. The antibody did not disrupt the ANGPTL8:ANGPTL3 complex, but came in close proximity to the LPL inhibitory motif in the N terminus of ANGPTL8. Collectively, these data show that ANGPTL8 has a functional LPL inhibitory motif, but only inhibits LPL and increases plasma TG levels in mice in the presence of ANGPTL3.
- Published
- 2017
- Full Text
- View/download PDF
8. Activin A more prominently regulates muscle mass in primates than does GDF8
- Author
-
Esther Latres, Jason Mastaitis, Wen Fury, Lawrence Miloscio, Jesus Trejos, Jeffrey Pangilinan, Haruka Okamoto, Katie Cavino, Erqian Na, Angelos Papatheodorou, Tobias Willer, Yu Bai, Jee Hae Kim, Ashique Rafique, Stephen Jaspers, Trevor Stitt, Andrew J. Murphy, George D. Yancopoulos, and Jesper Gromada
- Subjects
Science - Abstract
Inhibition of GDF8 increases muscle mass in mice, but is less effective in monkeys and humans. Here the authors show that activin A also inhibits muscle hypertrophy and that concomitant inhibition of activin A and GDF8 synergistically increases muscle mass in mice and non-human primates.
- Published
- 2017
- Full Text
- View/download PDF
9. PC1/3 Deficiency Impacts Pro-opiomelanocortin Processing in Human Embryonic Stem Cell-Derived Hypothalamic Neurons
- Author
-
Liheng Wang, Lina Sui, Sunil K. Panigrahi, Kana Meece, Yurong Xin, Jinrang Kim, Jesper Gromada, Claudia A. Doege, Sharon L. Wardlaw, Dieter Egli, and Rudolph L. Leibel
- Subjects
PC1/3 deficiency ,hESC ,hypothalamic neurons ,POMC processing ,obesity ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
We recently developed a technique for generating hypothalamic neurons from human pluripotent stem cells. Here, as proof of principle, we examine the use of these cells in modeling of a monogenic form of severe obesity: PCSK1 deficiency. The cognate enzyme, PC1/3, processes many prohormones in neuroendocrine and other tissues. We generated PCSK1 (PC1/3)-deficient human embryonic stem cell (hESC) lines using both short hairpin RNA and CRISPR-Cas9, and investigated pro-opiomelanocortin (POMC) processing using hESC-differentiated hypothalamic neurons. The increased levels of unprocessed POMC and the decreased ratios (relative to POMC) of processed POMC-derived peptides in both PCSK1 knockdown and knockout hESC-derived neurons phenocopied POMC processing reported in PC1/3-null mice and PC1/3-deficient patients. PC1/3 deficiency was associated with increased expression of melanocortin receptors and PRCP (prolylcarboxypeptidase, a catabolic enzyme for α-melanocyte stimulating hormone (αMSH)), and reduced adrenocorticotropic hormone secretion. We conclude that the obesity accompanying PCSK1 deficiency may not be primarily due to αMSH deficiency.
- Published
- 2017
- Full Text
- View/download PDF
10. Heterogeneity of human pancreatic β-cells
- Author
-
Giselle Dominguez-Gutierrez, Yurong Xin, and Jesper Gromada
- Subjects
Internal medicine ,RC31-1245 - Abstract
Background: Human pancreatic β-cells are heterogeneous. This has been known for a long time and is based on various functional and morphological readouts. β-Cell heterogeneity could reflect fixed subpopulations with distinct functions. However, recent pseudotime analysis of large-scale RNA sequencing data suggest that human β-cell subpopulations may rather reflect dynamic interchangeable states characterized by low expression of genes involved in the unfolded protein response (UPR) and low insulin gene expression, low UPR and high insulin expression or high UPR and low insulin expression. Scope of review: This review discusses findings obtained by single-cell RNA sequencing combined with pseudotime analysis that human β-cell heterogeneity represents dynamic interchangeable functional states. The physiological significance and potential implications of β-cell heterogeneity in the development and progression of diabetes is highlighted. Major conclusions: The existence of dynamic functional states allow β-cells to transition between periods of high insulin production and UPR-mediated stress recovery. The recovery state is important since proinsulin is a misfolding-prone protein, making its biosynthesis in the endoplasmic reticulum a stressful event. The transition of β-cells between dynamic states is likely controlled at multiple levels and influenced by the microenvironment within the pancreatic islets. Disturbances in the ability of the β-cells to transition between periods of high insulin biosynthesis and UPR-mediated stress recovery may contribute to diabetes development. Diabetes medications that restore the ability of the β-cells to transition between the functional states should be considered. Keywords: Human β-cell, Heterogeneity, Diabetes, Pancreatic islet, Insulin
- Published
- 2019
- Full Text
- View/download PDF
11. ANGPTL3 blockade with a human monoclonal antibody reduces plasma lipids in dyslipidemic mice and monkeys1[S]
- Author
-
Viktoria Gusarova, Corey A. Alexa, Yan Wang, Ashique Rafique, Jee Hae Kim, David Buckler, Ivory J. Mintah, Lisa M. Shihanian, Jonathan C. Cohen, Helen H. Hobbs, Yurong Xin, David M. Valenzuela, Andrew J. Murphy, George D. Yancopoulos, and Jesper Gromada
- Subjects
lipoprotein lipase ,endothelial lipase ,triglycerides ,cholesterol ,hyperlipidemia ,dyslipidemia ,Biochemistry ,QD415-436 - Abstract
Angiopoietin-like protein 3 (ANGPTL3) is a circulating protein synthesized exclusively in the liver that inhibits LPL and endothelial lipase (EL), enzymes that hydrolyze TGs and phospholipids in plasma lipoproteins. Here we describe the development and testing of a fully human monoclonal antibody (REGN1500) that binds ANGPTL3 with high affinity. REGN1500 reversed ANGPTL3-induced inhibition of LPL activity in vitro. Intravenous administration of REGN1500 to normolipidemic C57Bl/6 mice increased LPL activity and decreased plasma TG levels by ≥50%. Chronic administration of REGN1500 to dyslipidemic C57Bl/6 mice for 8 weeks reduced circulating plasma levels of TG, LDL-cholesterol (LDL-C), and HDL-cholesterol (HDL-C) without any changes in liver, adipose, or heart TG contents. Studies in EL knockout mice revealed that REGN1500 reduced serum HDL-C through an EL-dependent mechanism. Finally, administration of a single dose of REGN1500 to dyslipidemic cynomolgus monkeys caused a rapid and pronounced decrease in plasma TG, nonHDL-C, and HDL-C. REGN1500 normalized plasma TG levels even in monkeys with a baseline plasma TG greater than 400 mg/dl. Collectively, these data demonstrate that neutralization of ANGPTL3 using REGN1500 reduces plasma lipids in dyslipidemic mice and monkeys, and thus provides a potential therapeutic agent for treatment of patients with hyperlipidemia.
- Published
- 2015
- Full Text
- View/download PDF
12. Inactivation of ANGPTL3 reduces hepatic VLDL-triglyceride secretion1[S]
- Author
-
Yan Wang, Viktoria Gusarova, Serena Banfi, Jesper Gromada, Jonathan C. Cohen, and Helen H. Hobbs
- Subjects
cholesterol ,dyslipidemias ,lipase/endothelial ,lipase/lipoprotein ,angiopoietin-like protein 3 ,very low density lipoprotein ,Biochemistry ,QD415-436 - Abstract
Humans and mice lacking angiopoietin-like protein 3 (ANGPTL3) have pan-hypolipidemia. ANGPTL3 inhibits two intravascular lipases, LPL and endothelial lipase, and the low plasma TG and HDL-cholesterol levels in ANGPTL3 deficiency reflect increased activity of these enzymes. The mechanism responsible for the low LDL-cholesterol levels associated with ANGPTL3 deficiency is not known. Here we used an anti-ANGPTL3 monoclonal antibody (REGN1500) to inactivate ANGPTL3 in mice with genetic deficiencies in key proteins involved in clearance of ApoB-containing lipoproteins. REGN1500 treatment consistently reduced plasma cholesterol levels in mice in which Apoe, Ldlr, Lrp1, and Sdc1 were inactivated singly or in combination, but did not alter clearance of rabbit 125I-βVLDL or mouse 125I-LDL. Despite a 61% reduction in VLDL-TG production, VLDL-ApoB-100 production was unchanged in REGN1500-treated animals. Hepatic TG content, fatty acid synthesis, and fatty acid oxidation were similar in REGN1500 and control antibody-treated animals. Taken together, our findings indicate that inactivation of ANGPTL3 does not affect the number of ApoB-containing lipoproteins secreted by the liver but alters the particles that are made such that they are cleared more rapidly from the circulation via a noncanonical pathway(s). The increased clearance of lipolytic remnants results in decreased production of LDL in ANGPTL3-deficient animals.
- Published
- 2015
- Full Text
- View/download PDF
13. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance
- Author
-
Mark W. Sleeman, Lisa M. Shihanian, George D. Yancopoulos, Joseph Lee, Andrew J. Murphy, Jonathan Cohen, Jesper Gromada, Helen H. Hobbs, Corey A. Alexa-Braun, Viktoria Gusarova, Cristopher V. Van Hout, Sara Hamon, Poulabi Banerjee, Hye In Kim, Ivory J. Mintah, and Rene C. Adam
- Subjects
0301 basic medicine ,Endothelial lipase ,Very low-density lipoprotein ,Evinacumab ,Cholesterol, VLDL ,Familial hypercholesterolemia ,QD415-436 ,030204 cardiovascular system & hematology ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Mediator ,cardiovascular disease ,ANGPTL3 ,low density lipoprotein-cholesterol ,medicine ,Animals ,Humans ,Endothelium ,Research Articles ,Angiopoietin-Like Protein 3 ,familial hypercholesterolemia ,Catabolism ,Chemistry ,nutritional and metabolic diseases ,Cell Biology ,Cholesterol, LDL ,medicine.disease ,low density lipoprotein receptor ,Cell biology ,very low density lipoprotein ,030104 developmental biology ,Angiopoietin-like Proteins ,Receptors, LDL ,LDL receptor ,lipidomics ,lipids (amino acids, peptides, and proteins) ,atherosclerosis - Abstract
Angiopoietin-like protein (ANGPTL)3 regulates plasma lipids by inhibiting LPL and endothelial lipase (EL). ANGPTL3 inactivation lowers LDL-C independently of the classical LDLR-mediated pathway and represents a promising therapeutic approach for individuals with homozygous familial hypercholesterolemia due to LDLR mutations. Yet, how ANGPTL3 regulates LDL-C levels is unknown. Here, we demonstrate in hyperlipidemic humans and mice that ANGPTL3 controls VLDL catabolism upstream of LDL. Using kinetic, lipidomic, and biophysical studies, we show that ANGPTL3 inhibition reduces VLDL-lipid content and size, generating remnant particles that are efficiently removed from the circulation. This suggests that ANGPTL3 inhibition lowers LDL-C by limiting LDL particle production. Mechanistically, we discovered that EL is a key mediator of ANGPTL3's novel pathway. Our experiments revealed that, although dispensable in the presence of LDLR, EL-mediated processing of VLDL becomes critical for LDLR-independent particle clearance. In the absence of EL and LDLR, ANGPTL3 inhibition perturbed VLDL catabolism, promoted accumulation of atypical remnants, and failed to reduce LDL-C. Taken together, we uncover ANGPTL3 at the helm of a novel EL-dependent pathway that lowers LDL-C in the absence of LDLR.
- Published
- 2020
14. HNF1A deficiency causes reduced calcium levels, accumulation of abnormal insulin granules and uncoupled insulin to C-peptide secretion in a stem cell model of MODY3
- Author
-
Haoquan Zhao, Rudolph Leibel, Christopher N Goulbourne, Jose Oberholzer, Jaeyop Lee, Yufeng Shen, Yuan Xing, Damian J. Williams, Robin Goland, Jesper Gromada, Henry M. Colecraft, Charles A. LeDuc, Jacqueline Niu, Xiajuan Chen, Wendy K. Chung, Dieter Egli, Yong Wang, and Bryan J. González
- Subjects
endocrine system ,medicine.medical_specialty ,business.industry ,C-peptide ,Insulin ,medicine.medical_treatment ,chemistry.chemical_element ,Calcium ,HNF1A ,chemistry.chemical_compound ,Endocrinology ,Text mining ,chemistry ,Internal medicine ,medicine ,Secretion ,Stem cell ,business - Abstract
Mutations in HNF1A cause Maturity Onset Diabetes of the Young type 3 (MODY3), the most prevalent form of monogenic diabetes. Using stem cell-derived pancreatic endocrine cells from human embryonic stem cells (hESCs) with induced hypomorphic mutations in HNF1A, we show that HNF1A orchestrates a transcriptional program required for calcium-dependent insulin secretion. HNF1A-deficient β-cells display a reduction in CACNA1A and intracellular calcium levels, as well as impaired insulin granule exocytosis in association with SYT13 down-regulation. Knockout of CACNA1A and SYT13 reproduce the relevant phenotypes. Retention of insulin is associated with accumulation of enlarged secretory granules, and altered stoichiometry of secreted insulin to C-peptide. Glibenclamide, a sulfonylurea drug used in the treatment of MODY3 patients, increases intracellular calcium, and thereby restores C-peptide and insulin secretion to a normal ratio. While insulin secretion defects are constitutive in cells with complete HNF1A loss of function, β-cells from patients with heterozygous hypomorphic HNF1A mutations are initially normal, but lose the ability to secrete insulin and acquire abnormal stoichiometric secretion ratios, while gene corrected cells remain normal. Our studies provide the molecular basis for the treatment of MODY3 with sulfonylureas, and demonstrate promise for the use of cell therapies for MODY3.
- Published
- 2021
15. The Liver–α-Cell Axis and Type 2 Diabetes
- Author
-
Jesper Gromada, Hendrik Vilstrup, Katrine D. Galsgaard, Malte P. Suppli, Nicolai J. Wewer Albrechtsen, Marie Winther-Sørensen, Lina Janah, Jens Pedersen, Filip K. Knop, and Jens J. Holst
- Subjects
0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Incretin ,030209 endocrinology & metabolism ,Glucagon ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Insulin resistance ,Internal medicine ,Nonalcoholic fatty liver disease ,medicine ,Hyperinsulinemia ,Animals ,Humans ,Insulin ,Amino Acids ,business.industry ,Fatty liver ,nutritional and metabolic diseases ,medicine.disease ,Glucose ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,Liver ,Glucagon-Secreting Cells ,Insulin Resistance ,business ,Glucagon receptor ,hormones, hormone substitutes, and hormone antagonists ,Hyperglucagonemia - Abstract
Both type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD) strongly associate with increasing body mass index, and together these metabolic diseases affect millions of individuals. In patients with T2D, increased secretion of glucagon (hyperglucagonemia) contributes to diabetic hyperglycemia as proven by the significant lowering of fasting plasma glucose levels following glucagon receptor antagonist administration. Emerging data now indicate that the elevated plasma concentrations of glucagon may also be associated with hepatic steatosis and not necessarily with the presence or absence of T2D. Thus, fatty liver disease, most often secondary to overeating, may result in impaired amino acid turnover, leading to increased plasma concentrations of certain glucagonotropic amino acids (e.g., alanine). This, in turn, causes increased glucagon secretion that may help to restore amino acid turnover and ureagenesis, but it may eventually also lead to increased hepatic glucose production, a hallmark of T2D. Early experimental findings support the hypothesis that hepatic steatosis impairs glucagon’s actions on amino acid turnover and ureagenesis. Hepatic steatosis also impairs hepatic insulin sensitivity and clearance that, together with hyperglycemia and hyperaminoacidemia, lead to peripheral hyperinsulinemia; systemic hyperinsulinemia may itself contribute to worsen peripheral insulin resistance. Additionally, obesity is accompanied by an impaired incretin effect, causing meal-related glucose intolerance. Lipid-induced impairment of hepatic sensitivity, not only to insulin but potentially also to glucagon, resulting in both hyperinsulinemia and hyperglucagonemia, may therefore contribute to the development of T2D at least in a subset of individuals with NAFLD.
- Published
- 2019
16. Increased SLC38A4 Amino Acid Transporter Expression in Human Pancreatic α-Cells After Glucagon Receptor Inhibition
- Author
-
Guenter Kloeppel, Biin Sung, Jinrang Kim, Jesper Gromada, Bence Sipos, Haruka Okamoto, Katie Cavino, Yurong Xin, and Giselle Dominguez Gutierrez
- Subjects
Adult ,Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Amino Acid Transport System A ,Transplantation, Heterologous ,Islets of Langerhans Transplantation ,030209 endocrinology & metabolism ,Glucagon ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Receptors, Glucagon ,medicine ,Animals ,Humans ,Amino acid transporter ,Receptor ,Cell Proliferation ,chemistry.chemical_classification ,Hyperplasia ,Pancreatic islets ,Middle Aged ,Amino acid ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Glucagon-Secreting Cells ,Commentary ,Female ,Signal transduction ,Pancreas ,Glucagon receptor ,Signal Transduction - Abstract
Plasma amino acids and their transporters constitute an important part of the feedback loop between the liver and pancreatic α-cell function, and glucagon regulates hepatic amino acid turnover. Disruption of hepatic glucagon receptor action activates the loop and results in high plasma amino acids and hypersecretion of glucagon associated with α-cell hyperplasia. In the present study, we report a technique to rescue implanted human pancreatic islets from the mouse kidney capsule. Using this model, we have demonstrated that expression of the amino acid transporter SLC38A4 increases in α-cells after administration of a glucagon receptor blocking antibody. The increase in SLC38A4 expression and associated α-cell proliferation was dependent on mechanistic target of rapamycin pathway. We confirmed increased α-cell proliferation and expression of SLC38A4 in pancreas sections from patients with glucagon cell hyperplasia and neoplasia (GCHN) with loss-of-function mutations in the glucagon receptor. Collectively, using a technique to rescue implanted human islets from the kidney capsule in mice and pancreas sections from patients with GCHN, we found that expression of SLC38A4 was increased under conditions of disrupted glucagon receptor signaling. These data provide support for the existence of a liver-human α-cell endocrine feedback loop.
- Published
- 2019
17. Hepatic Glucagon Signaling Regulates PCSK9 and Low-Density Lipoprotein Cholesterol
- Author
-
John A. Zadroga, Wen Dai, Jesper Gromada, Haruka Okamoto, Lale Ozcan, Erika S. Wittchen, and Stefano Spolitu
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,Cholesterol ,PCSK9 ,Low density lipoprotein cholesterol ,030209 endocrinology & metabolism ,Lipid metabolism ,Glucagon ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Glucose homeostasis ,Cardiology and Cardiovascular Medicine ,Homeostasis ,Hormone - Abstract
Rationale: Glucagon is a key hormone that regulates the adaptive metabolic responses to fasting. In addition to maintaining glucose homeostasis, glucagon participates in the regulation of cholesterol metabolism; however, the molecular pathways underlying this effect are incompletely understood. Objective: We sought to determine the role of hepatic Gcgr (glucagon receptor) signaling in plasma cholesterol regulation and identify its underlying molecular mechanisms. Methods and Results: We show that Gcgr signaling plays an essential role in LDL-C (low-density lipoprotein cholesterol) homeostasis through regulating the PCSK9 (proprotein convertase subtilisin/kexin type 9) levels. Silencing of hepatic Gcgr or inhibition of glucagon action increased hepatic and plasma PCSK9 and resulted in lower LDLR (LDL receptor) protein and increased plasma LDL-C. Conversely, treatment of wild-type (WT) mice with glucagon lowered LDL-C levels, whereas this response was abrogated in Pcsk9 −/− and Ldlr −/− mice. Our gain- and loss-of-function studies identified Epac2 (exchange protein activated by cAMP-2) and Rap1 (Ras-related protein-1) as the downstream mediators of glucagon’s action on PCSK9 homeostasis. Moreover, mechanistic studies revealed that glucagon affected the half-life of PCSK9 protein without changing the level of its mRNA, indicating that Gcgr signaling regulates PCSK9 degradation. Conclusions: These findings provide novel insights into the molecular interplay between hepatic glucagon signaling and lipid metabolism and describe a new posttranscriptional mechanism of PCSK9 regulation.
- Published
- 2019
18. Reduced calcium levels and accumulation of abnormal insulin granules in stem cell models of HNF1A deficiency
- Author
-
Bryan J. González, Haoquan Zhao, Jacqueline Niu, Damian J. Williams, Jaeyop Lee, Chris N. Goulbourne, Yuan Xing, Yong Wang, Jose Oberholzer, Maria H. Blumenkrantz, Xiaojuan Chen, Charles A. LeDuc, Wendy K. Chung, Henry M. Colecraft, Jesper Gromada, Yufeng Shen, Robin S. Goland, Rudolph L. Leibel, and Dieter Egli
- Subjects
Synaptotagmins ,Diabetes Mellitus, Type 2 ,Insulin, Regular, Human ,Stem Cells ,Medicine (miscellaneous) ,Humans ,Insulin ,Calcium ,Hepatocyte Nuclear Factor 1-alpha ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Abstract
Mutations in HNF1A cause Maturity Onset Diabetes of the Young (HNF1A-MODY). To understand mechanisms of β-cell dysfunction, we generated stem cell-derived pancreatic endocrine cells with hypomorphic mutations in HNF1A. HNF1A-deficient β-cells display impaired basal and glucose stimulated-insulin secretion, reduced intracellular calcium levels in association with a reduction in CACNA1A expression, and accumulation of abnormal insulin granules in association with SYT13 down-regulation. Knockout of CACNA1A and SYT13 reproduce the relevant phenotypes. In HNF1A deficient β-cells, glibenclamide, a sulfonylurea drug used in the treatment of HNF1A-MODY patients, increases intracellular calcium, and restores insulin secretion. While insulin secretion defects are constitutive in β-cells null for HNF1A, β-cells heterozygous for hypomorphic HNF1A (R200Q) mutations lose the ability to secrete insulin gradually; this phenotype is prevented by correction of the mutation. Our studies illuminate the molecular basis for the efficacy of treatment of HNF1A-MODY with sulfonylureas, and suggest promise for the use of cell therapies.
- Published
- 2021
19. Human stem cell model of HNF1A deficiency shows uncoupled insulin to C-peptide secretion with accumulation of abnormal insulin granules
- Author
-
Robin Goland, Bryan J. González, Jesper Gromada, Yufeng Shen, Christopher N Goulbourne, Jacqueline Niu, Yuan Xing, Wendy K. Chung, Henry M. Colecraft, Dieter Egli, Xiaojuan Chen, Yong Wang, Jose Oberholzer, Charles A. LeDuc, Rudolph L. Leibel, Haoquan Zhao, Damian J. Williams, and Jaeyop Lee
- Subjects
endocrine system ,medicine.medical_specialty ,Chemistry ,Cellular differentiation ,Insulin ,medicine.medical_treatment ,Enteroendocrine cell ,Embryonic stem cell ,Alpha cell ,Endocrinology ,Internal medicine ,medicine ,Stem cell ,Beta cell ,Induced pluripotent stem cell - Abstract
Mutations in HNF1A cause Maturity Onset Diabetes of the Young type 3 (MODY3), the most prevalent form of monogenic diabetes. We generated stem cell-derived pancreatic endocrine cells from human embryonic stem cells (hESCs) with induced hypomorphic mutations in HNF1A. Using these cells, we show that HNF1A orchestrates a transcriptional program required for distinct aspects of β-cell fate and function. During islet cell differentiation, HNF1A deficiency biases islet endocrine cells towards an α-cell fate associated with PAX4 down-regulation. HNF1A- deficient β-cells display impaired basal and glucose stimulated-insulin secretion in association with a reduction in CACNA1A and intracellular calcium levels, and impaired insulin granule exocytosis in association with SYT13 down-regulation. Knockout of PAX4, CACNA1A and SYT13 reproduce the relevant phenotypes. Reduction of insulin secretion is associated with accumulation of enlarged secretory granules, and altered stoichiometry of secreted insulin to C-peptide. In HNF1A deficient β-cells, glibenclamide, a sulfonylurea drug used in the treatment of MODY3 patients, increases intracellular calcium to levels beyond those achieved by glucose, and restores C-peptide and insulin secretion to a normal stoichiometric ratio. To study HNF1A deficiency in the context of a human disease model, we also generated stem cell-derived pancreatic endocrine cells from two MODY3 patient’s induced pluripotent stem cells (iPSCs). While insulin secretion defects are constitutive in cells with complete HNF1A loss of function, β-cells heterozygous for hypomorphic HNF1A mutations are initially normal, but lose the ability to secrete insulin and acquire abnormal stoichiometric secretion ratios. Importantly, the defects observed in these stem cell models are also seen in circulating proportions of insulin:C-peptide in nine MODY3 patients.One sentence of summaryDeficiency of the transcription factor HNF1A biases islet endocrine cell fate towards α-cells, impairs intracellular calcium homeostasis and insulin exocytosis, alters the stoichiometry of insulin to C-peptide release, and leads to an accumulation of abnormal insulin secretory granules in β-cells.
- Published
- 2021
20. Glucagon Receptor Inhibition Reduces Hyperammonemia and Lethality in Male Mice with Urea Cycle Disorder
- Author
-
Katie Cavino, Jesper Gromada, Qi Su, Haruka Okamoto, Jinrang Kim, Erqian Na, Biin Sung, and Xiping Cheng
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Urea cycle disorder ,Ornithine transcarbamylase ,Ammonia homeostasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Glutaminase ,Ammonia ,Glutamate-Ammonia Ligase ,Glutamine synthetase ,Internal medicine ,medicine ,Receptors, Glucagon ,Animals ,Hyperammonemia ,Amino Acids ,Ornithine Carbamoyltransferase ,Chemistry ,Body Weight ,Antibodies, Monoclonal ,medicine.disease ,Ornithine Carbamoyltransferase Deficiency Disease ,030104 developmental biology ,Gene Expression Regulation ,Urea cycle ,Glucagon receptor ,030217 neurology & neurosurgery - Abstract
The liver plays a critical role in maintaining ammonia homeostasis. Urea cycle defects, liver injury, or failure and glutamine synthetase (GS) deficiency result in hyperammonemia, serious clinical conditions, and lethality. In this study we used a mouse model with a defect in the urea cycle enzyme ornithine transcarbamylase (Otcspf-ash) to test the hypothesis that glucagon receptor inhibition using a monoclonal blocking antibody will reduce the hyperammonemia and associated lethality induced by a high-protein diet, which exacerbates disease. We found reduced expression of glutaminase, which degrades glutamine and increased expression of GS in livers of Otcspf-ash mice treated with the glucagon receptor blocking antibody. The gene expression changes favor ammonia consumption and were accompanied by increased circulating glutamine levels and diminished hyperammonemia. Otcspf-ash mice treated with the glucagon receptor-blocking antibody gained lean and body mass and had increased survival. These data suggest that glucagon receptor inhibition using a monoclonal antibody could reduce the risk for hyperammonemia and other clinical manifestations of patients suffering from defects in the urea cycle, liver injury, or failure and GS deficiency.
- Published
- 2020
21. Discordance between GLP-1R gene and protein expression in mouse pancreatic islet cells
- Author
-
Yurong Xin, Sarah M. Gray, David A. D'Alessio, Megan E. Capozzi, Elizabeth C. Ross, Kimberley El, Berit Svendsen, Jenny Tong, Jesper Gromada, Peter Ravn, Jonathan E. Campbell, Bryanna M. Chazotte, and Kyle W. Sloop
- Subjects
0301 basic medicine ,endocrine system ,Incretin ,Gene Expression ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,Flow cytometry ,03 medical and health sciences ,Mice ,Insulin-Secreting Cells ,Gene expression ,medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,Gene ,Cells, Cultured ,Messenger RNA ,030102 biochemistry & molecular biology ,biology ,medicine.diagnostic_test ,digestive, oral, and skin physiology ,Cell Biology ,Cell sorting ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Metabolism ,biology.protein ,Antibody ,Single-Cell Analysis ,hormones, hormone substitutes, and hormone antagonists - Abstract
The insulinotropic actions of glucagon-like peptide 1 receptor (GLP-1R) in β-cells have made it a useful target to manage type 2 diabetes. Metabolic stress reduces β-cell sensitivity to GLP-1, yet the underlying mechanisms are unknown. We hypothesized that Glp1r expression is heterogeneous among β-cells and that metabolic stress decreases the number of GLP-1R–positive β-cells. Here, analyses of publicly available single-cell RNA-Seq sequencing (scRNASeq) data from mouse and human β-cells indicated that significant populations of β-cells do not express the Glp1r gene, supporting heterogeneous GLP-1R expression. To check these results, we used complementary approaches employing FACS coupled with quantitative RT-PCR, a validated GLP-1R antibody, and flow cytometry to quantify GLP-1R promoter activity, gene expression, and protein expression in mouse α-, β-, and δ-cells. Experiments with Glp1r reporter mice and a validated GLP-1R antibody indicated that >90% of the β-cells are GLP-1R positive, contradicting the findings with the scRNASeq data. α-cells did not express Glp1r mRNA and δ-cells expressed Glp1r mRNA but not protein. We also examined the expression patterns of GLP-1R in mouse models of metabolic stress. Multiparous female mice had significantly decreased β-cell Glp1r expression, but no reduction in GLP-1R protein levels or GLP-1R–mediated insulin secretion. These findings suggest caution in interpreting the results of scRNASeq for low-abundance transcripts such as the incretin receptors and indicate that GLP-1R is widely expressed in β-cells, absent in α-cells, and expressed at the mRNA, but not protein, level in δ-cells.
- Published
- 2020
22. Characterization of glucose-stimulated insulin release protocols in african green monkeys (Chlorocebus aethiops)
- Author
-
Shervin Liddie, Matthew S Lawrence, Haruka Okamoto, and Jesper Gromada
- Subjects
Male ,medicine.medical_specialty ,endocrine system diseases ,Intravenous Glucose Tolerance ,nonhuman primates ,medicine.medical_treatment ,oral glucose tolerance test ,Chlorocebus aethiops ,Glucose infusion ,graded glucose infusion ,vervet ,Internal medicine ,Diabetes mellitus ,Insulin Secretion ,medicine ,Animals ,Oral glucose tolerance ,Insulin secretion ,plasma insulin ,diabetes ,General Veterinary ,biology ,business.industry ,Insulin ,aging ,nutritional and metabolic diseases ,Original Articles ,Glucose Tolerance Test ,insulinopenic ,biology.organism_classification ,medicine.disease ,African green monkey ,Endocrinology ,Original Article ,Female ,Animal Science and Zoology ,African Green Monkey ,business ,intravenous glucose tolerance test - Abstract
Background Management of diabetes remains a major health and economic challenge, demanding test systems in which to develop new therapies. These studies assessed different methodologies for determining glucose tolerance in green monkeys. Methods Twenty‐eight African green monkeys between 4 and 24 years old underwent single or repeat intravenous glucose tolerance testing (IVGTT), oral glucose tolerance testing (OGTT), and/or graded glucose infusion testing. Results Geriatric monkeys exhibited glucose intolerance with impaired glucose‐stimulated insulin secretion following IVGTT. Repeat IVGTT and OGTT assessments were inconsistent. Monkeys with low glucose‐stimulated insulin secretion after graded glucose infusion exhibited elevated blood glucose levels. Conclusion IVGTT and graded glucose infusion protocols revealed differences in glucose tolerance among green monkeys at single time points, including age‐dependent differences suggestive of shifts in pancreatic beta‐cell functional capacity, but care should be applied to study design and the interpretation of data in the setting of longitudinal studies.
- Published
- 2018
23. The α-cell in diabetes mellitus
- Author
-
Guy A. Rutter, Jesper Gromada, and Pauline Chabosseau
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Cell ,Disease ,Sensitivity and Specificity ,03 medical and health sciences ,Endocrinology ,Insulin-Secreting Cells ,Internal medicine ,Diabetes mellitus ,medicine ,Humans ,Insulin ,Type 1 diabetes ,business.industry ,Role ,Glucagon secretion ,Type 2 Diabetes Mellitus ,Glucagon ,medicine.disease ,Glutamine ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,Glucagon-Secreting Cells ,Female ,business - Abstract
Findings from the past 10 years have placed the glucagon-secreting pancreatic α-cell centre stage in the development of diabetes mellitus, a disease affecting almost one in every ten adults worldwide. Glucagon secretion is reduced in patients with type 1 diabetes mellitus, increasing the risk of insulin-induced hypoglycaemia, but is enhanced in type 2 diabetes mellitus, exacerbating the effects of diminished insulin release and action on blood levels of glucose. A better understanding of the mechanisms underlying these changes is therefore an important goal. RNA sequencing reveals that, despite their opposing roles in the control of blood levels of glucose, α-cells and β-cells have remarkably similar patterns of gene expression. This similarity might explain the fairly facile interconversion between these cells and the ability of the α-cell compartment to serve as a source of new β-cells in models of extreme β-cell loss that mimic type 1 diabetes mellitus. Emerging data suggest that GABA might facilitate this interconversion, whereas the amino acid glutamine serves as a liver-derived factor to promote α-cell replication and maintenance of α-cell mass. Here, we survey these developments and their therapeutic implications for patients with diabetes mellitus.
- Published
- 2018
24. Gene Signature of Proliferating Human Pancreatic α Cells
- Author
-
Christina Adler, Ann-Hwee Lee, Jesper Gromada, Giselle Dominguez Gutierrez, Jinrang Kim, Min Ni, Andrew J. Murphy, Haruka Okamoto, Yurong Xin, and George D. Yancopoulos
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Cell division ,030209 endocrinology & metabolism ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Humans ,RNA, Messenger ,Gene ,Cell Proliferation ,Sequence Analysis, RNA ,Chemistry ,Cell Cycle ,RNA ,Middle Aged ,Cell cycle ,Gene signature ,Cell biology ,030104 developmental biology ,Glucagon-Secreting Cells ,Female ,Single-Cell Analysis ,Transcriptome - Abstract
Pancreatic α cells proliferate at a low rate, and little is known about the control of this process. Here we report the characterization of human α cells by large-scale, single-cell RNA sequencing coupled with pseudotime ordering. We identified two large subpopulations and a smaller cluster of proliferating α cells with increased expression of genes involved in cell-cycle regulation. The proliferating α cells were differentiated, had normal levels of GCG expression, and showed no signs of cellular stress. Proliferating α cells were detected in both the G1S and G2M phases of the cell cycle. Human α cells proliferate at a fivefold higher rate than human β cells and express lower levels of the cell-cycle inhibitors CDKN1A and CDKN1C. Collectively, this study provides the gene signatures of human α cells and the genes involved in their cell division. The lower expression of two cell-cycle inhibitors in human α cells could account for their higher rate of proliferation compared with their insulin-producing counterparts.
- Published
- 2018
25. Genetic inactivation of ANGPTL4 improves glucose homeostasis and is associated with reduced risk of diabetes
- Author
-
Teresa Tusié-Luna, Svati H. Shah, Wen-Jane Lee, Amit Khera, Gonçalo R. Abecasis, Olle Melander, Alan R. Shuldiner, Connor A. Emdin, Kari Stefansson, Jesper Gromada, Andrew P. Morris, Lee-Ming Chuang, Omri Gottesman, Lars G. Fritsche, Pradeep Natarajan, Marju Orho-Melander, Daniel F. Gudbjartsson, Anubha Mahajan, Marylyn D. Ritchie, William E. Kraus, Tooraj Mirshahi, Colm O'Dushlaine, Jason Flannick, Nicholas J. Wareham, Anne Tybjærg-Hansen, Anders Berg Wulff, Rashmi B. Prasad, Aris Baras, Jonas B. Nielsen, Valgerdur Steinthorsdottir, Yii-Der Ida Chen, Jerome I. Rotter, Lukas Habegger, Samantha N. Fetterolf, David Altshuler, Om Prakash Dwivedi, Tanya M. Teslovich, Cristen J. Willer, Luca A. Lotta, Andrew J. Murphy, Joseph B. Leader, Cristopher V. Van Hout, Christopher D. Still, Ola Hansson, David Birtwell, Alexander Lopez, Daniel J. Rader, John D. Overton, Anthony Marcketta, Patrick Sulem, Peter N. Benotti, Jose C. Florez, Lydia Coulter Kwee, David J. Carey, Oddgeir L. Holmen, Kristian Hveem, Leif Groop, Sekar Kathiresan, Viktoria Gusarova, Unnur Thorsteinsdottir, Cassandra M. Hartle, Uyenlinh L. Mirshahi, H. Lester Kirchner, Shannon Bruse, Robert A. Scott, Michael F. Murray, Marketa Sjögren, Jeffrey G. Reid, Aeron Small, Børge G. Nordestgaard, Amr H. Wardeh, Chad M. Brummett, Mark I. McCarthy, Frederick E. Dewey, David H. Ledbetter, John Penn, Ingrid B. Borecki, Scott M. Damrauer, Hilma Holm, Michael Boehnke, George D. Yancopoulos, Institute for Molecular Medicine Finland, University of Helsinki, Centre of Excellence in Complex Disease Genetics, and HUS Abdominal Center
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,Insulin Resistance/genetics ,General Physics and Astronomy ,Type 2 diabetes ,030204 cardiovascular system & hematology ,Inbred C57BL ,Cardiovascular ,HAN CHINESE ,Whole Exome Sequencing ,Mice ,0302 clinical medicine ,Risk Factors ,ANGPTL4 ,Homeostasis ,Glucose homeostasis ,lcsh:Science ,Mice, Knockout ,Lipoprotein lipase ,Multidisciplinary ,Diabetes ,Lipoprotein Lipase/metabolism ,REMNANT CHOLESTEROL ,ADIPOSE-TISSUE ,Female ,Type 2 ,Heterozygote ,medicine.medical_specialty ,Knockout ,Science ,LIPOPROTEIN-LIPASE ,HEART-DISEASE ,Diabetes Mellitus, Type 2/etiology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Angiopoietin-like 4 Protein/deficiency ,03 medical and health sciences ,Internal medicine ,Diabetes mellitus ,Exome Sequencing ,Diabetes Mellitus ,Genetics ,medicine ,Angiopoietin-Like Protein 4 ,Animals ,Humans ,Gene Silencing ,GENOME-WIDE ASSOCIATION ,Metabolic and endocrine ,Genetic Association Studies ,CHINESE POPULATION ,Blood Glucose/metabolism ,PLASMA-LIPIDS ,business.industry ,Case-control study ,Genetic Variation ,General Chemistry ,Odds ratio ,Atherosclerosis ,medicine.disease ,Mice, Inbred C57BL ,Lipoprotein Lipase ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,Amino Acid Substitution ,Case-Control Studies ,lcsh:Q ,3111 Biomedicine ,ANGIOPOIETIN-LIKE PROTEIN-4 ,Insulin Resistance ,business - Abstract
Angiopoietin-like 4 (ANGPTL4) is an endogenous inhibitor of lipoprotein lipase that modulates lipid levels, coronary atherosclerosis risk, and nutrient partitioning. We hypothesize that loss of ANGPTL4 function might improve glucose homeostasis and decrease risk of type 2 diabetes (T2D). We investigate protein-altering variants in ANGPTL4 among 58,124 participants in the DiscovEHR human genetics study, with follow-up studies in 82,766 T2D cases and 498,761 controls. Carriers of p.E40K, a variant that abolishes ANGPTL4 ability to inhibit lipoprotein lipase, have lower odds of T2D (odds ratio 0.89, 95% confidence interval 0.85–0.92, p = 6.3 × 10−10), lower fasting glucose, and greater insulin sensitivity. Predicted loss-of-function variants are associated with lower odds of T2D among 32,015 cases and 84,006 controls (odds ratio 0.71, 95% confidence interval 0.49–0.99, p = 0.041). Functional studies in Angptl4-deficient mice confirm improved insulin sensitivity and glucose homeostasis. In conclusion, genetic inactivation of ANGPTL4 is associated with improved glucose homeostasis and reduced risk of T2D., Genetic variation in ANGPTL4 is associated with lipid traits. Here, the authors find that predicted loss-of-function variants in ANGPTL4 are associated with glucose homeostasis and reduced risk of type 2 diabetes and that Angptl4−/− mice on a high-fat diet show improved insulin sensitivity.
- Published
- 2018
26. Glucagon contributes to liver zonation
- Author
-
Sun Y. Kim, Haruka Okamoto, Jesper Gromada, George D. Yancopoulos, Andrew J. Murphy, Yurong Xin, and Xiping Cheng
- Subjects
0301 basic medicine ,Conservation of Natural Resources ,Multidisciplinary ,Physiology ,Wnt signaling pathway ,glucagon receptor ,Biology ,Biological Sciences ,Glucagon ,Cell biology ,03 medical and health sciences ,Metabolic pathway ,Wnt ,030104 developmental biology ,PNAS Plus ,glucagon ,Gene expression ,liver zonation ,Signal transduction ,City Planning ,Receptor ,Gene ,Glucagon receptor - Abstract
Significance The lobules are the functional units of the liver. They consist of 15–25 layers of hepatocytes with specialized metabolic functions and gene expression patterns relative to their position along the lobule, a phenomenon referred to as metabolic zonation. The Wnt/β-catenin pathway regulates hepatocyte function but how the zonation is controlled to meet the metabolic demands of the liver is unclear. Glucagon regulates hepatic function. We now demonstrate that glucagon contributes to liver zonation by interacting and opposing the actions of the Wnt/β-catenin pathway., Liver zonation characterizes the separation of metabolic pathways along the lobules and is required for optimal function. Wnt/β-catenin signaling controls metabolic zonation by activating genes in the perivenous hepatocytes, while suppressing genes in the periportal counterparts. We now demonstrate that glucagon opposes the actions of Wnt/β-catenin signaling on gene expression and metabolic zonation pattern. The effects were more pronounced in the periportal hepatocytes where 28% of all genes were activated by glucagon and inhibited by Wnt/β-catenin. The glucagon and Wnt/β-catenin receptors and their signaling pathways are uniformly distributed in periportal and perivenous hepatocytes and the expression is not regulated by the opposing signal. Collectively, our results show that glucagon controls gene expression and metabolic zonation in the liver through a counterplay with the Wnt/β-catenin signaling pathway.
- Published
- 2018
27. Increased thermogenesis by a noncanonical pathway in ANGPTL3/8-deficient mice
- Author
-
Helen H. Hobbs, Jonathan C. Cohen, Jesper Gromada, Serena Banfi, and Viktoria Gusarova
- Subjects
0301 basic medicine ,Hyperthermia ,Male ,medicine.medical_specialty ,Medical Sciences ,Adipose Tissue, White ,Adipose tissue ,White adipose tissue ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Oxygen Consumption ,ANGPTL3 ,Adipose Tissue, Brown ,Angiopoietin-Like Protein 8 ,Internal medicine ,Brown adipose tissue ,medicine ,Animals ,Beta oxidation ,Fatty acid synthesis ,Triglycerides ,Angiopoietin-Like Protein 3 ,2. Zero hunger ,Mice, Knockout ,Multidisciplinary ,Triglyceride ,Thermogenesis ,Biological Sciences ,medicine.disease ,hyperthermia ,Lipid Metabolism ,Dietary Fats ,ANGPTL8 ,adipose tissue ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Angiopoietin-like Proteins ,chemistry ,PNAS Plus ,13. Climate action ,Receptors, Adrenergic, beta-3 ,Female - Abstract
Significance White adipose tissue (WAT) serves as an energy reservoir during fasting and is replenished with fatty acids from circulating triglycerides upon refeeding. We showed previously that postprandial partitioning of fatty acids between oxidative and storage tissues is mediated by angiopoietin-like proteins 3 (A3) and 8 (A8). Here, we show that disruption of both Angptl3 and Angptl8 in mice causes striking alterations in energy metabolism: reduced fat mass, hyperthermia, increased metabolic rate, and beiging of subcutaneous WAT. The hypermetabolic features of the A3−/−A8−/− mice were most pronounced in fed animals, and attenuated with fasting and β3-adrenergic receptor blockade. These data indicate that A3 and A8 promote efficient energy utilization by tissues and limit the increase in energy expenditure associated with food intake., Dietary triglyceride (TG) is the most efficient energy substrate. It is processed and stored at substantially lower metabolic cost than is protein or carbohydrate. In fed animals, circulating TGs are preferentially routed for storage to white adipose tissue (WAT) by angiopoietin-like proteins 3 (A3) and 8 (A8). Here, we show that mice lacking A3 and A8 (A3−/−A8−/− mice) have decreased fat mass and a striking increase in temperature (+1 °C) in the fed (but not fasted) state, without alterations in food intake or physical activity. Subcutaneous WAT (WAT-SQ) from these animals had morphologic and metabolic changes characteristic of beiging. O2 consumption rates (OCRs) and expression of genes involved in both fatty acid synthesis and fatty acid oxidation were increased in WAT-SQ of A3−/−A8−/− mice, but not in their epididymal or brown adipose tissue (BAT). The hyperthermic response to feeding was blocked by maintaining A3−/−A8−/− mice at thermoneutrality or by treating with a β3-adrenergic receptor (AR) antagonist. To determine if sympathetic stimulation was sufficient to increase body temperature in A3−/−A8−/− mice, WT and A3−/−A8−/− animals were maintained at thermoneutrality and then treated with a β3-AR agonist; treatment induced hyperthermia in A3−/−A8−/−, but not WT, mice. Antibody-mediated inactivation of both circulating A3 and A8 induced hyperthermia in WT mice. Together, these data indicate that A3 and A8 are essential for efficient storage of dietary TG and that disruption of these genes increases feeding-induced thermogenesis and energy utilization.
- Published
- 2018
28. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease
- Author
-
Alex Lopez, Jerome I. Rotter, Yii-Der Ida Chen, Lukas Habegger, Anders Berg Wulff, Richard L Dunbar, John Penn, Anita M. van den Hoek, An Zhao, Omri Gottesman, Cristopher V. Van Hout, Scott M. Damrauer, Tanya M. Teslovich, Aurelie Bouzelmat, Daniel J. Rader, Poulabi Banerjee, David J. Carey, Viktoria Gusarova, Shane McCarthy, Frederick E. Dewey, Kae-Woei Liang, David H. Ledbetter, Gary Herman, Rick Zhang, Alan R. Shuldiner, Svati H. Shah, Claudia Schurmann, Neil Stahl, Marylyn D. Ritchie, Sara Hamon, Colm O'Dushlaine, Ingrid B. Borecki, Daniel A. Gipe, Jesper Gromada, Hans M.G. Princen, Børge G. Nordestgaard, H. Lester Kirchner, Michael F. Murray, Shannon Bruse, Aris Baras, Jeffrey G. Reid, Wayne H-H Sheu, William E. Kraus, Xiuqing Guo, Aeron Small, Brad Shumel, William J. Sasiela, Anne Tybjærg-Hansen, Andrew J. Murphy, Joseph B. Leader, John D. Overton, Robert Pordy, Scott Mellis, Weiping Shao, George D. Yancopoulos, Hayes Dansky, and I-Te Lee
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Evinacumab ,Population ,030204 cardiovascular system & hematology ,Pharmacology ,Coronary artery disease ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,ANGPTL3 ,Medicine ,education ,education.field_of_study ,biology ,business.industry ,Cholesterol ,General Medicine ,medicine.disease ,Human genetics ,030104 developmental biology ,Endocrinology ,chemistry ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Antibody ,business ,Lipoprotein - Abstract
BackgroundLoss-of-function variants in the angiopoietin-like 3 gene (ANGPTL3) have been associated with decreased plasma levels of triglycerides, low-density lipoprotein (LDL) cholesterol, and high-density lipoprotein (HDL) cholesterol. It is not known whether such variants or therapeutic antagonism of ANGPTL3 are associated with a reduced risk of atherosclerotic cardiovascular disease. MethodsWe sequenced the exons of ANGPTL3 in 58,335 participants in the DiscovEHR human genetics study. We performed tests of association for loss-of-function variants in ANGPTL3 with lipid levels and with coronary artery disease in 13,102 case patients and 40,430 controls from the DiscovEHR study, with follow-up studies involving 23,317 case patients and 107,166 controls from four population studies. We also tested the effects of a human monoclonal antibody, evinacumab, against Angptl3 in dyslipidemic mice and against ANGPTL3 in healthy human volunteers with elevated levels of triglycerides or LDL cholesterol. ResultsIn th...
- Published
- 2017
29. Inflammatory Ly6Chi monocytes and their conversion to M2 macrophages drive atherosclerosis regression
- Author
-
Yuliya Vengrenyuk, Karishma Rahman, Natasha M. Girgis, P'ng Loke, Stephen A. Ramsey, Viktoria Gusarova, Noemi Rotllan Vila, Edward A. Fisher, Kathryn J. Moore, Jianhua Liu, Ada Weinstock, and Jesper Gromada
- Subjects
Male ,0301 basic medicine ,Receptors, CCR5 ,Receptors, CCR2 ,CX3C Chemokine Receptor 1 ,Hyperlipidemias ,Inflammation ,030204 cardiovascular system & hematology ,Monocytes ,Mice ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,CX3CR1 ,Animals ,Medicine ,Macrophage ,Cell Lineage ,Receptor ,Aorta ,STAT6 ,Mice, Knockout ,Sequence Analysis, RNA ,business.industry ,Macrophages ,Monocyte ,General Medicine ,Atherosclerosis ,Phenotype ,Plaque, Atherosclerotic ,Mice, Inbred C57BL ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Female ,Receptors, Chemokine ,medicine.symptom ,STAT6 Transcription Factor ,business ,Research Article - Abstract
Atherosclerosis is a chronic inflammatory disease, and developing therapies to promote its regression is an important clinical goal. We previously established that atherosclerosis regression is characterized by an overall decrease in plaque macrophages and enrichment in markers of alternatively activated M2 macrophages. We have now investigated the origin and functional requirement for M2 macrophages in regression in normolipidemic mice that received transplants of atherosclerotic aortic segments. We compared plaque regression in WT normolipidemic recipients and those deficient in chemokine receptors necessary to recruit inflammatory Ly6Chi (Ccr2–/– or Cx3cr1–/–) or patrolling Ly6Clo (Ccr5–/–) monocytes. Atherosclerotic plaques transplanted into WT or Ccr5–/– recipients showed reduced macrophage content and increased M2 markers consistent with plaque regression, whereas plaques transplanted into Ccr2–/– or Cx3cr1–/– recipients lacked this regression signature. The requirement of recipient Ly6Chi monocyte recruitment was confirmed in cell trafficking studies. Fate-mapping and single-cell RNA sequencing studies also showed that M2-like macrophages were derived from newly recruited monocytes. Furthermore, we used recipient mice deficient in STAT6 to demonstrate a requirement for this critical component of M2 polarization in atherosclerosis regression. Collectively, these results suggest that continued recruitment of Ly6Chi inflammatory monocytes and their STAT6-dependent polarization to the M2 state are required for resolution of atherosclerotic inflammation and plaque regression.
- Published
- 2017
30. PC1/3 Deficiency Impacts Pro-opiomelanocortin Processing in Human Embryonic Stem Cell-Derived Hypothalamic Neurons
- Author
-
Yurong Xin, Jinrang Kim, Jesper Gromada, Liheng Wang, Dieter Egli, Lina Sui, Rudolph L. Leibel, Claudia A. Doege, Sharon L. Wardlaw, Sunil K Panigrahi, and Kana Meece
- Subjects
0301 basic medicine ,obesity ,Pro-Opiomelanocortin ,Human Embryonic Stem Cells ,Gene Expression ,Apoptosis ,Biochemistry ,Small hairpin RNA ,Mice ,Receptor ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cells, Cultured ,Neurons ,Gene knockdown ,lcsh:R5-920 ,Pyramidal Cells ,Cell Differentiation ,Endoplasmic Reticulum Stress ,Immunohistochemistry ,Proprotein Convertase 1 ,Gene Knockdown Techniques ,Gene Targeting ,Melanocortin ,lcsh:Medicine (General) ,hormones, hormone substitutes, and hormone antagonists ,medicine.medical_specialty ,endocrine system ,Biology ,Article ,POMC processing ,03 medical and health sciences ,Adrenocorticotropic Hormone ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,PC1/3 deficiency ,Catabolism ,Cell Biology ,Embryonic stem cell ,030104 developmental biology ,Endocrinology ,nervous system ,lcsh:Biology (General) ,alpha-MSH ,hESC ,Mutation ,Proteolysis ,CRISPR-Cas Systems ,hypothalamic neurons ,Developmental Biology ,Hormone - Abstract
Summary We recently developed a technique for generating hypothalamic neurons from human pluripotent stem cells. Here, as proof of principle, we examine the use of these cells in modeling of a monogenic form of severe obesity: PCSK1 deficiency. The cognate enzyme, PC1/3, processes many prohormones in neuroendocrine and other tissues. We generated PCSK1 (PC1/3)-deficient human embryonic stem cell (hESC) lines using both short hairpin RNA and CRISPR-Cas9, and investigated pro-opiomelanocortin (POMC) processing using hESC-differentiated hypothalamic neurons. The increased levels of unprocessed POMC and the decreased ratios (relative to POMC) of processed POMC-derived peptides in both PCSK1 knockdown and knockout hESC-derived neurons phenocopied POMC processing reported in PC1/3-null mice and PC1/3-deficient patients. PC1/3 deficiency was associated with increased expression of melanocortin receptors and PRCP (prolylcarboxypeptidase, a catabolic enzyme for α-melanocyte stimulating hormone (αMSH)), and reduced adrenocorticotropic hormone secretion. We conclude that the obesity accompanying PCSK1 deficiency may not be primarily due to αMSH deficiency., Highlights • Stem cell-derived hypothalamic neurons are used to study human obesity • shRNA and CRISPR-Cas9 were used to generate models of PCSK1 deficiency • PC1/3 deficiency impaired POMC processing in arcuate-like neurons • Adaptive changes occurred in “downstream” POMC processing enzymes, Leibel and colleagues use hESC-derived arcuate hypothalamic neurons to examine the neuro-molecular physiology of the hyperphagic obesity that accompanies hypomorphic mutations of PCSK1 in human subjects. They find that reduced proconvertase (PC1/3) activity impairs processing of the propeptide, POMC, and increases the expression of downstream processing enzymes without affecting the final production of αMSH. Decreased levels of the POMC product, ACTH, may contribute to the hyperphagia.
- Published
- 2017
- Full Text
- View/download PDF
31. Glucagon receptor inhibition normalizes blood glucose in severe insulin-resistant mice
- Author
-
Sun Y. Kim, Elizabeth A. Krumm, Andrew J. Murphy, Katie Cavino, Haruka Okamoto, Erqian Na, Jesper Gromada, Xiping Cheng, and George D. Yancopoulos
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Multidisciplinary ,biology ,business.industry ,medicine.drug_class ,Antagonist ,030209 endocrinology & metabolism ,Biological Sciences ,Monoclonal antibody ,medicine.disease ,03 medical and health sciences ,Insulin receptor ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,biology.protein ,Medicine ,Antibody ,business ,Antagonism ,Glucagon receptor - Abstract
Inactivating mutations in the insulin receptor results in extreme insulin resistance. The resulting hyperglycemia is very difficult to treat, and patients are at risk for early morbidity and mortality from complications of diabetes. We used the insulin receptor antagonist S961 to induce severe insulin resistance, hyperglycemia, and ketonemia in mice. Using this model, we show that glucagon receptor (GCGR) inhibition with a monoclonal antibody normalized blood glucose and β-hydroxybutyrate levels. Insulin receptor antagonism increased pancreatic β-cell mass threefold. Normalization of blood glucose levels with GCGR-blocking antibody unexpectedly doubled β-cell mass relative to that observed with S961 alone and 5.8-fold over control. GCGR antibody blockage expanded α-cell mass 5.7-fold, and S961 had no additional effects. Collectively, these data show that GCGR antibody inhibition represents a potential therapeutic option for treatment of patients with extreme insulin-resistance syndromes.
- Published
- 2017
32. Heterogeneity of human pancreatic β-cells
- Author
-
Yurong Xin, Giselle Dominguez-Gutierrez, and Jesper Gromada
- Subjects
0301 basic medicine ,lcsh:Internal medicine ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Review ,Biology ,Pancreatic islet ,03 medical and health sciences ,0302 clinical medicine ,Insulin-Secreting Cells ,Gene expression ,medicine ,Diabetes Mellitus ,Humans ,Insulin ,lcsh:RC31-1245 ,Molecular Biology ,Gene ,Proinsulin ,Human β-cell ,Sequence Analysis, RNA ,Pancreatic islets ,Endoplasmic reticulum ,Diabetes ,RNA ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Unfolded protein response ,Unfolded Protein Response ,Heterogeneity - Abstract
Background: Human pancreatic β-cells are heterogeneous. This has been known for a long time and is based on various functional and morphological readouts. β-Cell heterogeneity could reflect fixed subpopulations with distinct functions. However, recent pseudotime analysis of large-scale RNA sequencing data suggest that human β-cell subpopulations may rather reflect dynamic interchangeable states characterized by low expression of genes involved in the unfolded protein response (UPR) and low insulin gene expression, low UPR and high insulin expression or high UPR and low insulin expression. Scope of review: This review discusses findings obtained by single-cell RNA sequencing combined with pseudotime analysis that human β-cell heterogeneity represents dynamic interchangeable functional states. The physiological significance and potential implications of β-cell heterogeneity in the development and progression of diabetes is highlighted. Major conclusions: The existence of dynamic functional states allow β-cells to transition between periods of high insulin production and UPR-mediated stress recovery. The recovery state is important since proinsulin is a misfolding-prone protein, making its biosynthesis in the endoplasmic reticulum a stressful event. The transition of β-cells between dynamic states is likely controlled at multiple levels and influenced by the microenvironment within the pancreatic islets. Disturbances in the ability of the β-cells to transition between periods of high insulin biosynthesis and UPR-mediated stress recovery may contribute to diabetes development. Diabetes medications that restore the ability of the β-cells to transition between the functional states should be considered. Keywords: Human β-cell, Heterogeneity, Diabetes, Pancreatic islet, Insulin
- Published
- 2019
33. Clinical and Molecular Prevalence of Lipodystrophy in an Unascertained Large Clinical Care Cohort
- Author
-
David J. Carey, Aris Baras, H. Lester Kirchner, Wenzhen Ge, Ryan Colonie, Jesper Gromada, Jeffrey Staples, Ashish Yadav, Joseph B. Leader, Alan R. Shuldiner, Claudia Gonzaga-Jauregui, Judith Altarejos, Michael F. Murray, Jeffrey G. Reid, Cristopher V. Van Hout, John D. Overton, Omri Gottesman, and Steve Gao
- Subjects
0301 basic medicine ,Adult ,Male ,Pediatrics ,medicine.medical_specialty ,Dunnigan familial partial lipodystrophy ,Lipodystrophy ,Endocrinology, Diabetes and Metabolism ,Population ,030209 endocrinology & metabolism ,Disease ,LMNA ,03 medical and health sciences ,0302 clinical medicine ,Internal Medicine ,Medicine ,Electronic Health Records ,Humans ,Genetic Predisposition to Disease ,education ,education.field_of_study ,business.industry ,Partial Lipodystrophy ,Genomics ,medicine.disease ,United States ,030104 developmental biology ,Population Surveillance ,Cohort ,Female ,Diagnosis code ,business - Abstract
Lipodystrophies are a group of disorders characterized by absence or loss of adipose tissue and abnormal fat distribution, commonly accompanied by metabolic dysregulation. Although considered rare disorders, their prevalence in the general population is not well understood. We aimed to evaluate the clinical and genetic prevalence of lipodystrophy disorders in a large clinical care cohort. We interrogated the electronic health record (EHR) information of >1.3 million adults from the Geisinger Health System for lipodystrophy diagnostic codes. We estimate a clinical prevalence of disease of 1 in 20,000 individuals. We performed genetic analyses in individuals with available genomic data to identify variants associated with inherited lipodystrophies and examined their EHR for comorbidities associated with lipodystrophy. We identified 16 individuals carrying the p.R482Q pathogenic variant in LMNA associated with Dunnigan familial partial lipodystrophy. Four had a clinical diagnosis of lipodystrophy, whereas the remaining had no documented clinical diagnosis despite having accompanying metabolic abnormalities. We observed a lipodystrophy-associated variant carrier frequency of 1 in 3,082 individuals in our cohort with substantial burden of metabolic dysregulation. We estimate a genetic prevalence of disease of ∼1 in 7,000 in the general population. Partial lipodystrophy is an underdiagnosed condition. and its prevalence, as defined molecularly, is higher than previously reported. Genetically guided stratification of patients with common metabolic disorders, like diabetes and dyslipidemia, is an important step toward precision medicine.
- Published
- 2019
34. Loss of ZnT8 function protects against diabetes by enhanced insulin secretion
- Author
-
Maddalena Trombetta, Fernando Abaitua, Isabella Artner, Nicola L. Beer, Ulrika Krus, Reshma Ramracheya, Jesper Gromada, Jason Flannick, Philipp Kramer, Martijn van de Bunt, Mark I. McCarthy, Deepak Jain, Patrik Rorsman, Enzo Bonora, Rebecca Cheung, Julia Brosnan, Ioannis Spiliotis, Anu Suoranta, Pauline Chabosseau, Antje Grotz, Ann Marie Richard, Claes B. Wolheim, Ola Hansson, Riccardo C. Bonadonna, Anna L. Gloyn, Leif Groop, Om Prakash Dwivedi, Mikko Lehtovirta, Anthony Payne, Timo Otonkoski, Vikash Chandra, L Sarelin, Nicole A.J. Krentz, Sandra Kleiner, Benjamin Davies, Soren K. Thomsen, Benoit Hastoy, Guy A. Rutter, Tiinamaija Tuomi, Daniel Gomez, Benoite Champon, Jens O. Lagerstedt, Emma Ahlqvist, Aris Baras, Daniela Moralli, Rashmi B. Prasad, and Andria Theodoulou
- Subjects
Adult ,Male ,medicine.medical_specialty ,endocrine system ,endocrine system diseases ,Adolescent ,Genotype ,Induced Pluripotent Stem Cells ,Type 2 diabetes ,Zinc Transporter 8 ,Biology ,Article ,Aged ,Diabetes Mellitus, Type 2 ,Female ,Glucose ,Humans ,Islets of Langerhans ,Middle Aged ,Young Adult ,Insulin Secretion ,03 medical and health sciences ,0302 clinical medicine ,Diabetes mellitus ,Internal medicine ,Diabetes Mellitus ,Genetics ,medicine ,Glucose homeostasis ,Allele ,11 Medical and Health Sciences ,030304 developmental biology ,Proinsulin ,0303 health sciences ,SLC30A8 ,Glucagon secretion ,06 Biological Sciences ,medicine.disease ,Endocrinology ,biology.protein ,Type 2 ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
A rare loss-of-function allele p.Arg138* in SLC30A8 encoding the zinc transporter 8 (ZnT8), which is enriched in Western Finland, protects against type 2 diabetes (T2D). We recruited relatives of the identified carriers and showed that protection was associated with better insulin secretion due to enhanced glucose responsiveness and proinsulin conversion, particularly when compared with individuals matched for the genotype of a common T2D-risk allele in SLC30A8, p.Arg325. In genome-edited human induced pluripotent stem cell (iPSC)-derived β-like cells, we establish that the p.Arg138* allele results in reduced SLC30A8 expression due to haploinsufficiency. In human β cells, loss of SLC30A8 leads to increased glucose responsiveness and reduced KATP channel function similar to isolated islets from carriers of the T2D-protective allele p.Trp325. These data position ZnT8 as an appealing target for treatment aimed at maintaining insulin secretion capacity in T2D.
- Published
- 2019
- Full Text
- View/download PDF
35. RNA Sequencing of Single Human Islet Cells Reveals Type 2 Diabetes Genes
- Author
-
Yurong Xin, Jinrang Kim, Min Ni, Haruka Okamoto, Jesper Gromada, Calvin Lin, George D. Yancopoulos, Yi Wei, Christina Adler, and Andrew J. Murphy
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,030209 endocrinology & metabolism ,Type 2 diabetes ,Biology ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Single-cell analysis ,Internal medicine ,medicine ,Animals ,Humans ,Pancreatic polypeptide ,Molecular Biology ,Gene ,geography ,geography.geographical_feature_category ,Sequence Analysis, RNA ,Gene Expression Profiling ,RNA ,Cell Biology ,medicine.disease ,Islet ,Cell biology ,Gene expression profiling ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,Single-Cell Analysis ,Signal Transduction - Abstract
Pancreatic islet cells are critical for maintaining normal blood glucose levels, and their malfunction underlies diabetes development and progression. We used single-cell RNA sequencing to determine the transcriptomes of 1,492 human pancreatic α, β, δ, and PP cells from non-diabetic and type 2 diabetes organ donors. We identified cell-type-specific genes and pathways as well as 245 genes with disturbed expression in type 2 diabetes. Importantly, 92% of the genes have not previously been associated with islet cell function or growth. Comparison of gene profiles in mouse and human α and β cells revealed species-specific expression. All data are available for online browsing and download and will hopefully serve as a resource for the islet research community.
- Published
- 2016
36. Glucagon orchestrates stress‐induced hyperglycaemia
- Author
-
Jesper Gromada, George D. Yancopoulos, and Joyce Harp
- Subjects
insulin ,endocrine system ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Review Article ,030204 cardiovascular system & hematology ,Glucagon ,Glucagon-Like Peptide-1 Receptor ,stress‐induced hyperglycaemia ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Stress, Physiological ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,Humans ,critical illness ,Receptor ,Review Articles ,business.industry ,Mortality rate ,Insulin ,Stress induced ,medicine.disease ,Diabetes Mellitus, Type 1 ,Diabetes Mellitus, Type 2 ,Hyperglycemia ,ICU ,business ,Glucagon receptor ,Hormone - Abstract
Hyperglycaemia is commonly observed on admission and during hospitalization for medical illness, traumatic injury, burn and surgical intervention. This transient hyperglycaemia is referred to as stress‐induced hyperglycaemia (SIH) and frequently occurs in individuals without a history of diabetes. SIH has many of the same underlying hormonal disturbances as diabetes mellitus, specifically absolute or relative insulin deficiency and glucagon excess. SIH has the added features of elevated blood levels of catecholamines and cortisol, which are not typically present in people with diabetes who are not acutely ill. The seriousness of SIH is highlighted by its greater morbidity and mortality rates compared with those of hospitalized patients with normal glucose levels, and this increased risk is particularly high in those without pre‐existing diabetes. Insulin is the treatment standard for SIH, but new therapies that reduce glucose variability and hypoglycaemia are desired. In the present review, we focus on the key role of glucagon in SIH and discuss the potential use of glucagon receptor blockers and glucagon‐like peptide‐1 receptor agonists in SIH to achieve target glucose control.
- Published
- 2016
37. Mechano-signalling pathways in an experimental intensive critical illness myopathy model
- Author
-
Roberta Sartori, Stefano Gastaldello, Jesper Gromada, Wen Fury, Rebeca Corpeno Kalamgi, Lars Larsson, Jorge L. Ruas, Vicente Martínez-Redondo, Denis C. Guttridge, Yu Bai, Marco Sandri, and Heba Salah
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Critical Illness Myopathy ,Physiology ,Biology ,Muscle mass ,Intensive care unit ,Rats sprague dawley ,law.invention ,Sprague dawley ,03 medical and health sciences ,030104 developmental biology ,law ,Critical illness ,medicine ,Signalling pathways ,Neuroscience - Abstract
KEY POINTS: Using an experimental rat intensive care unit (ICU) model, not limited by early mortality, we have previously shown that passive mechanical loading attenuates the loss of muscle mass an ...
- Published
- 2016
38. Gene Signature of the Human Pancreatic ε Cell
- Author
-
Jesper Gromada, Jingjing Niu, Giselle Dominguez Gutierrez, Yueming Ding, Andrew J. Murphy, Sarah M. Gray, Min Ni, Jinrang Kim, Jenny Tong, Ann-Hwee Lee, Yi Wei, Yurong Xin, and Christina Adler
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,Cell type ,Cell ,Enteroendocrine cell ,Cell Count ,Cell Separation ,Biology ,Transcriptome ,03 medical and health sciences ,Islets of Langerhans ,Endocrinology ,Internal medicine ,medicine ,Humans ,Transcription factor ,Pancreas ,Research Articles ,Cells, Cultured ,Pancreatic islets ,Gene Expression Profiling ,Gene signature ,Microarray Analysis ,Ghrelin ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Signal Transduction - Abstract
The ghrelin-producing ε cell represents the fifth endocrine cell type in human pancreatic islets. The abundance of ε cells in adult pancreas is extremely low, which has hampered the investigation on the molecular pathways regulating the development and the function of this cell type. In this study, we explored the molecular features defining the function of pancreatic ε cells isolated from adult nondiabetic donors using single-cell RNA sequencing technology. We focus on transcription factors, cell surface receptors, and genes involved in metabolic pathways that contribute to regulation of cellular function. Furthermore, the genes that separate ε cells from the other islet endocrine cell types are presented. This study expands prior knowledge about the genes important for ε cell functioning during development and provides a resource to interrogate the transcriptome of this rare human islet cell type.
- Published
- 2018
39. Mice harboring the human SLC30A8 R138X loss-of-function mutation have increased insulin secretory capacity
- Author
-
Ramandeep Bhavsar, Jesper Gromada, Katie Cavino, Erqian Na, Giselle Dominguez-Gutierrez, George D. Yancopoulos, Ming Hu, Bezawit Megra, Yurong Xin, Brian Zambrowicz, Pauline Chabosseau, Rojas Jose F, Sandra Kleiner, Guy A. Rutter, Andrew J. Murphy, Gaelle Carrat, Daniel Gomez, Medical Research Council (MRC), and Wellcome Trust
- Subjects
0301 basic medicine ,MECHANISM ,Blood Glucose ,Male ,insulin secretion ,Physiology ,medicine.medical_treatment ,Type 2 diabetes ,Mice ,Loss of Function Mutation ,Insulin-Secreting Cells ,EXOCYTOSIS ,Glucose homeostasis ,Insulin ,Gene Knock-In Techniques ,Mice, Knockout ,Multidisciplinary ,biology ,SLC30A8 ,Chemistry ,Biological Sciences ,ZINC TRANSPORTER ZNT8 ,Multidisciplinary Sciences ,medicine.anatomical_structure ,PNAS Plus ,Zinc Transporter 8 ,genetic mutation ,Science & Technology - Other Topics ,EXPRESSION ,medicine.medical_specialty ,zinc transporter ,Carbohydrate metabolism ,03 medical and health sciences ,BETA-CELLS ,Internal medicine ,MD Multidisciplinary ,medicine ,Animals ,Humans ,Alleles ,Science & Technology ,Pancreatic islets ,medicine.disease ,Receptor, Insulin ,Mice, Inbred C57BL ,Insulin receptor ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Glucose ,Diabetes Mellitus, Type 2 ,Hyperglycemia ,biology.protein ,pancreatic beta cell ,GLUCOSE-HOMEOSTASIS ,Peptides - Abstract
Significance The zinc transporter SLC30A8 is primarily expressed in islets of the endocrine pancreas. Human SLC30A8 loss-of-function mutations protect against type 2 diabetes. However, Slc30a8 knockout mice do not show this protection. We have generated a mouse model mimicking a common protective human SLC30A8 loss-of-function allele. This mouse model shows a beneficial effect of loss of SLC30A8 function on β-cell biology. In particular, mice carrying the protective R138X allele have an increased capacity to secrete insulin in high-glucose conditions. Understanding the signaling mechanisms regulating insulin secretion in the R138X mice could provide novel insights into β-cell biology, and may lead to the identification of therapeutic targets for the treatment of diabetes., SLC30A8 encodes a zinc transporter that is primarily expressed in the pancreatic islets of Langerhans. In β-cells it transports zinc into insulin-containing secretory granules. Loss-of-function (LOF) mutations in SLC30A8 protect against type 2 diabetes in humans. In this study, we generated a knockin mouse model carrying one of the most common human LOF mutations for SLC30A8, R138X. The R138X mice had normal body weight, glucose tolerance, and pancreatic β-cell mass. Interestingly, in hyperglycemic conditions induced by the insulin receptor antagonist S961, the R138X mice showed a 50% increase in insulin secretion. This effect was not associated with enhanced β-cell proliferation or mass. Our data suggest that the SLC30A8 R138X LOF mutation may exert beneficial effects on glucose metabolism by increasing the capacity of β-cells to secrete insulin under hyperglycemic conditions.
- Published
- 2018
40. Pseudotime Ordering of Single Human β-Cells Reveals States of Insulin Production and Unfolded Protein Response
- Author
-
Yurong Xin, Haruka Okamoto, Jinrang Kim, Jesper Gromada, Min Ni, Andrew J. Murphy, Christina Adler, Giselle Dominguez Gutierrez, Ann-Hwee Lee, and George D. Yancopoulos
- Subjects
0301 basic medicine ,endocrine system ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,03 medical and health sciences ,0302 clinical medicine ,Insulin-Secreting Cells ,Gene expression ,Databases, Genetic ,Insulin Secretion ,Internal Medicine ,medicine ,Humans ,Insulin ,RNA, Messenger ,Transcription factor ,Cells, Cultured ,In Situ Hybridization, Fluorescence ,Proinsulin ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Principal Component Analysis ,Chemistry ,Sequence Analysis, RNA ,Endoplasmic reticulum ,Gene Expression Profiling ,Nucleotide Mapping ,Endoplasmic Reticulum Stress ,Cell biology ,Gene expression profiling ,Kinetics ,030104 developmental biology ,Gene Expression Regulation ,Multigene Family ,Unfolded protein response ,Unfolded Protein Response ,Single-Cell Analysis ,Biomarkers ,Transcription Factors - Abstract
Proinsulin is a misfolding-prone protein, making its biosynthesis in the endoplasmic reticulum (ER) a stressful event. Pancreatic β-cells overcome ER stress by activating the unfolded protein response (UPR) and reducing insulin production. This suggests that β-cells transition between periods of high insulin biosynthesis and UPR-mediated recovery from cellular stress. We now report the pseudotime ordering of single β-cells from humans without diabetes detected by large-scale RNA sequencing. We identified major states with 1) low UPR and low insulin gene expression, 2) low UPR and high insulin gene expression, or 3) high UPR and low insulin gene expression. The latter state was enriched for proliferating cells. Stressed human β-cells do not dedifferentiate and show little propensity for apoptosis. These data suggest that human β-cells transition between states with high rates of biosynthesis to fulfill the body’s insulin requirements to maintain normal blood glucose levels and UPR-mediated recovery from ER stress due to high insulin production.
- Published
- 2018
41. A Protein-Truncating HSD17B13 Variant and Protection from Chronic Liver Disease
- Author
-
Shane McCarthy, Jonathan C. Cohen, Frederick E. Dewey, Claudia Schurmann, Matthew D. Still, Panayiotis Stevis, Xin Chu, Daniel J. Rader, David J. Carey, Alan R. Shuldiner, Noura S. Abul-Husn, Semanti Mukherjee, Jonathan S. Packer, Xiping Cheng, Ann Stepanchick, Brian Zambrowicz, Helen H. Hobbs, John Penn, Uyenlinh L. Mirshahi, Scott M. Damrauer, Ingrid B. Borecki, Yurong Xin, G. Craig Wood, Jesper Gromada, Suganthi Balasubramanian, Tanya M. Teslovich, Andrew J. Murphy, Erin D. Fuller, Christopher D. Still, Tooraj Mirshahi, Jonathan Z. Luo, John D. Overton, George D. Yancopoulos, Yashu Liu, Alexander H. Li, Aeron Small, Omri Gottesman, Julia Kozlitina, Jeffrey G. Reid, Stefan Stender, David Esopi, William C. Olson, Michael Feldman, Colm O'Dushlaine, Alexander E. Lopez, Nehal Gosalia, Sun Y. Kim, and Aris Baras
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,17-Hydroxysteroid Dehydrogenases ,Genotype ,Chronic liver disease ,Gastroenterology ,Article ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Loss of Function Mutation ,Internal medicine ,Exome Sequencing ,Medicine ,Humans ,Exome ,Genetic Predisposition to Disease ,Aspartate Aminotransferases ,Exome sequencing ,biology ,business.industry ,Sequence Analysis, RNA ,Liver Diseases ,Fatty liver ,Genetic Variation ,Alanine Transaminase ,General Medicine ,medicine.disease ,Human genetics ,Fatty Liver ,030104 developmental biology ,Alanine transaminase ,Liver ,Chronic Disease ,biology.protein ,Disease Progression ,Linear Models ,030211 gastroenterology & hepatology ,Female ,business ,Biomarkers ,TM6SF2 - Abstract
BACKGROUND: Elucidation of the genetic factors underlying chronic liver disease may reveal new therapeutic targets. METHODS: We used exome sequence data and electronic health records from 46,544 participants in the DiscovEHR human genetics study to identify genetic variants associated with serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Variants that were replicated in three additional cohorts (12,527 persons) were evaluated for association with clinical diagnoses of chronic liver disease in DiscovEHR study participants and two independent cohorts (total of 37,173 persons) and with histopathological severity of liver disease in 2391 human liver samples. RESULTS: A splice variant (rs72613567:TA) in HSD17B13, encoding the hepatic lipid droplet protein hydroxysteroid 17-beta dehydrogenase 13, was found to be associated with reduced levels of ALT (P=4.20×10(−12)) and AST (P=6.2×10(−10)). Among DiscovEHR study participants, this variant was found to be associated with a reduced risk of alcoholic liver disease (by 42% [95% confidence interval {CI}, 20 to 58] among heterozygotes and by 53% [95% CI, 3 to 77] among homozygotes), nonalcoholic liver disease (by 17% [95% CI, 8 to 25] among heterozygotes and by 30% [95% CI, 13 to 43] among homozygotes), alcoholic cirrhosis (by 42% [95% CI, 14 to 61] among heterozygotes and by 73% [95% CI, 15 to 91] among homozygotes), and nonalcoholic cirrhosis (by 26% [95% CI, 7 to 40] among heterozygotes and by 49% [95% CI, 15 to 69] among homozygotes). Associations were confirmed in two independent cohorts. The rs72613567:TA variant was associated with a reduced risk of nonalcoholic steatohepatitis, but not steatosis, in human liver samples. The rs72613567:TA variant mitigated liver injury associated with the risk-increasing PNPLA3 p.I148M allele and resulted in an unstable and truncated protein with reduced enzymatic activity. CONCLUSIONS: A loss-of-function variant in HSD17B13 is associated with a reduced risk of chronic liver disease and of progression from steatosis to steatohepatitis. (Funded by Regeneron Pharmaceuticals and others.)
- Published
- 2018
42. Preemptive Activation of the Integrated Stress Response Protects Mice From Diet-Induced Obesity and Insulin Resistance by Fibroblast Growth Factor 21 Induction
- Author
-
Corey D. Holman, Jesper Gromada, Curtis J. Bare, David E. Cohen, Ann-Hwee Lee, Xu Xu, Christopher Steven Krumm, and Jae-Seon So
- Subjects
0301 basic medicine ,medicine.medical_specialty ,FGF21 ,Eukaryotic Initiation Factor-2 ,Cellular homeostasis ,Diet, High-Fat ,Article ,03 medical and health sciences ,Mice ,Insulin resistance ,Metabolic Diseases ,Stress, Physiological ,Internal medicine ,Protein Phosphatase 1 ,medicine ,Integrated stress response ,Glucose homeostasis ,Animals ,Homeostasis ,Adipocytes, Beige ,Obesity ,Adiposity ,Mice, Knockout ,Hepatology ,Chemistry ,medicine.disease ,Activating Transcription Factor 4 ,Diet ,Fatty Liver ,Fibroblast Growth Factors ,030104 developmental biology ,Endocrinology ,Phosphorylation ,Phosphatase complex ,Steatohepatitis ,Insulin Resistance ,Energy Metabolism - Abstract
The integrated stress response (ISR) is a signaling system in which the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by stress-specific kinases and the subsequent activation of activation transcription factor (ATF) 4 help restore cellular homeostasis following exposure to environmental stresses. ISR activation has been observed in metabolic diseases including hepatic steatosis, steatohepatitis and insulin resistance, but it remains unclear whether ISR contributes to the disease pathogenesis, or represents an innate defense mechanism against metabolic stresses. Constitutive repressor of eIF2α phosphorylation (CReP) is a critical regulatory subunit of the eIF2α phosphatase complex. Here we show that CReP ablation causes constitutive eIF2α phosphorylation in the liver, which leads to activation of the ATF4 transcriptional program including increased Fibroblast growth factor 21 (FGF21) production. Liver-specific CReP knockout (CReP(LKO)) mice exhibited marked browning of white adipose tissue, increased energy expenditure and insulin sensitivity in a FGF21-dependent manner. Furthermore, CReP(LKO) mice were protected from high fat diet (HFD)-induced obesity, hepatic steatosis, and insulin resistance. Acute CReP ablation in the liver of HFD-induced obese mice also reduced adiposity and improved glucose homeostasis. CONCLUSION: These data suggest that CReP abundance is a critical determinant for eIF2α phosphorylation and ensuing ISR activation in the liver. Constitutive ISR activation in the liver induces FGF21 and confers protection from high-fat diet (HFD)-induced adiposity, insulin resistance and hepatic steatosis in mice. Augmenting hepatic ISR may represent a novel therapeutic approach to treat metabolic disorders.
- Published
- 2018
43. Inositol hexakisphosphate kinase 1 is a metabolic sensor in pancreatic beta-cells
- Author
-
Christopher Illies, Subu Surendran Rajasekaran, Jesper Gromada, Gian-Carlo Gaboardi, Sung Ho Ryu, Martin Köhler, Stephen B. Shears, Joilson O. Martins, Sabrina de Souza Ferreira, Eduardo Lima Nolasco, Karen Tiago dos Santos, Jaeyoon Kim, Elisabetta Daré, Per Olof Berggren, Chunfang Gu, and Christopher J. Barker
- Subjects
0301 basic medicine ,INSULINA ,medicine.medical_treatment ,Inositol Phosphates ,T2D, type 2 diabetes ,ATP/ADP ,Carbohydrate metabolism ,Diphosphoinositol pentakisphosphate/ IP7 ,Exocytosis ,Article ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adenosine Triphosphate ,Insulin-Secreting Cells ,medicine ,Inositol hexakisphosphate kinase 1 ,Animals ,Humans ,Secretion ,Pancreatic beta cell ,IP6K, inositol hexakisphosphate kinase ,Phosphotransferases (Phosphate Group Acceptor) ,Inositol Hexakisphosphate Kinase 1 ,Kinase ,Chemistry ,Insulin ,Insulin secretion ,Type 2 diabetes ,Cell Biology ,Metabolism ,3. Good health ,Cell biology ,Adenosine Diphosphate ,Mice, Inbred C57BL ,030104 developmental biology ,Glucose ,Diabetes Mellitus, Type 2 ,IP7, di-phosphoinositol pentakisphosphate ,Gene Knockdown Techniques ,ATP–ADP translocase ,030217 neurology & neurosurgery - Abstract
Diphosphoinositol pentakisphosphate (IP7) is critical for the exocytotic capacity of the pancreatic β-cell, but its regulation by the primary instigator of β-cell exocytosis, glucose, is unknown. The high Km for ATP of the IP7-generating enzymes, the inositol hexakisphosphate kinases (IP6K1 and 2) suggests that these enzymes might serve as metabolic sensors in insulin secreting β-cells and act as translators of disrupted metabolism in diabetes. We investigated this hypothesis and now show that glucose stimulation, which increases the ATP/ADP ratio, leads to an early rise in IP7 concentration in β-cells. RNAi mediated knock down of the IP6K1 isoform inhibits both glucose-mediated increase in IP7 and first phase insulin secretion, demonstrating that IP6K1 integrates glucose metabolism and insulin exocytosis. In diabetic mouse islets the deranged ATP/ADP levels under both basal and glucose-stimulated conditions are mirrored in both disrupted IP7 generation and insulin release. Thus the unique metabolic sensing properties of IP6K1 guarantees appropriate concentrations of IP7 and thereby both correct basal insulin secretion and intact first phase insulin release. In addition, our data suggest that a specific cell signaling defect, namely, inappropriate IP7 generation may be an essential convergence point integrating multiple metabolic defects into the commonly observed phenotype in diabetes., Graphical abstract Unlabelled Image, Highlights • Glucose increases IP7 levels transiently through IP6K1 in pancreatic β-cells. • IP6K1 decodes glucose-driven increases in ATP/ADP ratio into 1st phase insulin release. • IP7 production and insulin release mirror perturbed metabolism in diabetic islets. • IP6K1 acts as a β-cell metabolic sensor under normal and pathological conditions.
- Published
- 2018
44. Hepatic ANGPTL3 regulates adipose tissue energy homeostasis
- Author
-
Helen H. Hobbs, Jonathan C. Cohen, Michael G. Levin, Viktoria Gusarova, Markey C. McNutt, Jesper Gromada, William L. Holland, Serena Banfi, and Yan Wang
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,Adipose Tissue, White ,Glucose uptake ,Adipose tissue ,White adipose tissue ,Biology ,Mice ,chemistry.chemical_compound ,Internal medicine ,Brown adipose tissue ,Cyclic AMP ,medicine ,Animals ,Homeostasis ,Insulin ,Tissue Distribution ,Triglycerides ,Angiopoietin-Like Protein 3 ,Mice, Knockout ,Lipoprotein lipase ,Multidisciplinary ,Triglyceride ,Fatty Acids ,nutritional and metabolic diseases ,Biological Sciences ,Hormones ,Mice, Inbred C57BL ,Lipoprotein Lipase ,Angiopoietin-like Proteins ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,Liver ,chemistry ,Lipogenesis ,Body Composition ,Female ,lipids (amino acids, peptides, and proteins) ,Angiopoietins ,Signal Transduction ,Lipoprotein - Abstract
Angiopoietin-like protein 3 (ANGPTL3) is a circulating inhibitor of lipoprotein and endothelial lipase whose physiological function has remained obscure. Here we show that ANGPTL3 plays a major role in promoting uptake of circulating very low density lipoprotein-triglycerides (VLDL-TGs) into white adipose tissue (WAT) rather than oxidative tissues (skeletal muscle, heart brown adipose tissue) in the fed state. This conclusion emerged from studies of Angptl3(-/-) mice. Whereas feeding increased VLDL-TG uptake into WAT eightfold in wild-type mice, no increase occurred in fed Angptl3(-/-) animals. Despite the reduction in delivery to and retention of TG in WAT, fat mass was largely preserved by a compensatory increase in de novo lipogenesis in Angptl3(-/-) mice. Glucose uptake into WAT was increased 10-fold in KO mice, and tracer studies revealed increased conversion of glucose to fatty acids in WAT but not liver. It is likely that the increased uptake of glucose into WAT explains the increased insulin sensitivity associated with inactivation of ANGPTL3. The beneficial effects of ANGPTL3 deficiency on both glucose and lipoprotein metabolism make it an attractive therapeutic target.
- Published
- 2015
45. ANGPTL3 blockade with a human monoclonal antibody reduces plasma lipids in dyslipidemic mice and monkeys
- Author
-
Jee Hae Kim, Lisa M. Shihanian, Buckler David R, Jonathan C. Cohen, Andrew J. Murphy, Viktoria Gusarova, Yan Wang, Ashique Rafique, Yurong Xin, David M. Valenzuela, Corey A. Alexa, George D. Yancopoulos, Helen H. Hobbs, Ivory J. Mintah, and Jesper Gromada
- Subjects
Male ,Endothelial lipase ,medicine.medical_specialty ,Evinacumab ,Adipose tissue ,Biochemistry ,Mice ,chemistry.chemical_compound ,Endocrinology ,ANGPTL3 ,Internal medicine ,Hyperlipidemia ,medicine ,Animals ,Humans ,Triglycerides ,Research Articles ,Angiopoietin-Like Protein 3 ,Dyslipidemias ,Lipoprotein lipase ,Chemistry ,Cholesterol ,Cholesterol, HDL ,Antibodies, Monoclonal ,Cholesterol, LDL ,Lipase ,Cell Biology ,medicine.disease ,Lipids ,Rats ,Lipoprotein Lipase ,Macaca fascicularis ,Angiopoietin-like Proteins ,Knockout mouse ,lipids (amino acids, peptides, and proteins) ,Angiopoietins - Abstract
Angiopoietin-like protein 3 (ANGPTL3) is a circulating protein synthesized exclusively in the liver that inhibits LPL and endothelial lipase (EL), enzymes that hydrolyze TGs and phospholipids in plasma lipoproteins. Here we describe the development and testing of a fully human monoclonal antibody (REGN1500) that binds ANGPTL3 with high affinity. REGN1500 reversed ANGPTL3-induced inhibition of LPL activity in vitro. Intravenous administration of REGN1500 to normolipidemic C57Bl/6 mice increased LPL activity and decreased plasma TG levels by ≥50%. Chronic administration of REGN1500 to dyslipidemic C57Bl/6 mice for 8 weeks reduced circulating plasma levels of TG, LDL-cholesterol (LDL-C), and HDL-cholesterol (HDL-C) without any changes in liver, adipose, or heart TG contents. Studies in EL knockout mice revealed that REGN1500 reduced serum HDL-C through an EL-dependent mechanism. Finally, administration of a single dose of REGN1500 to dyslipidemic cynomolgus monkeys caused a rapid and pronounced decrease in plasma TG, nonHDL-C, and HDL-C. REGN1500 normalized plasma TG levels even in monkeys with a baseline plasma TG greater than 400 mg/dl. Collectively, these data demonstrate that neutralization of ANGPTL3 using REGN1500 reduces plasma lipids in dyslipidemic mice and monkeys, and thus provides a potential therapeutic agent for treatment of patients with hyperlipidemia.
- Published
- 2015
46. Inactivation of ANGPTL3 reduces hepatic VLDL-triglyceride secretion
- Author
-
Jonathan C. Cohen, Viktoria Gusarova, Serena Banfi, Jesper Gromada, Helen H. Hobbs, and Yan Wang
- Subjects
Male ,Apolipoprotein E ,Endothelial lipase ,medicine.medical_specialty ,Very low-density lipoprotein ,Evinacumab ,Lipoproteins, VLDL ,Biology ,Biochemistry ,Mice ,chemistry.chemical_compound ,Apolipoproteins E ,Endocrinology ,Internal medicine ,medicine ,Animals ,Humans ,Gene Silencing ,Beta oxidation ,Triglycerides ,Research Articles ,Fatty acid synthesis ,Angiopoietin-Like Protein 3 ,Cholesterol ,Tumor Suppressor Proteins ,Antibodies, Monoclonal ,Cell Biology ,Angiopoietin-like Proteins ,Liver ,Receptors, LDL ,chemistry ,LDL receptor ,lipids (amino acids, peptides, and proteins) ,Rabbits ,Syndecan-1 ,Angiopoietins ,Low Density Lipoprotein Receptor-Related Protein-1 - Abstract
Humans and mice lacking angiopoietin-like protein 3 (ANGPTL3) have pan-hypolipidemia. ANGPTL3 inhibits two intravascular lipases, LPL and endothelial lipase, and the low plasma TG and HDL-cholesterol levels in ANGPTL3 deficiency reflect increased activity of these enzymes. The mechanism responsible for the low LDL-cholesterol levels associated with ANGPTL3 deficiency is not known. Here we used an anti-ANGPTL3 monoclonal antibody (REGN1500) to inactivate ANGPTL3 in mice with genetic deficiencies in key proteins involved in clearance of ApoB-containing lipoproteins. REGN1500 treatment consistently reduced plasma cholesterol levels in mice in which Apoe, Ldlr, Lrp1, and Sdc1 were inactivated singly or in combination, but did not alter clearance of rabbit (125)I-βVLDL or mouse (125)I-LDL. Despite a 61% reduction in VLDL-TG production, VLDL-ApoB-100 production was unchanged in REGN1500-treated animals. Hepatic TG content, fatty acid synthesis, and fatty acid oxidation were similar in REGN1500 and control antibody-treated animals. Taken together, our findings indicate that inactivation of ANGPTL3 does not affect the number of ApoB-containing lipoproteins secreted by the liver but alters the particles that are made such that they are cleared more rapidly from the circulation via a noncanonical pathway(s). The increased clearance of lipolytic remnants results in decreased production of LDL in ANGPTL3-deficient animals.
- Published
- 2015
47. Amino Acid Transporter Slc38a5 Controls Glucagon Receptor Inhibition-Induced Pancreatic α Cell Hyperplasia in Mice
- Author
-
Guillermo Anguiano, Brian Zambrowicz, Haruka Okamoto, Wen Hong Li, Jesper Gromada, Jinrang Kim, Shiuhwei Chen, Erqian Na, Qing Liu, George D. Yancopoulos, Joseph Lee, Yurong Xin, Zhi Jiang Huang, Roger H Unger, Katie Cavino, Yan Xu, Rachid Hamid, and Andrew J. Murphy
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Physiology ,Glucagon ,Article ,03 medical and health sciences ,Mice ,Internal medicine ,medicine ,Receptors, Glucagon ,Glucose homeostasis ,Animals ,Amino acid transporter ,Molecular Biology ,Glucagon-like peptide 1 receptor ,chemistry.chemical_classification ,Mice, Knockout ,Mice, Inbred ICR ,Hyperplasia ,Chemistry ,Cell Biology ,medicine.disease ,Amino acid ,030104 developmental biology ,Endocrinology ,Amino Acid Transport Systems, Neutral ,Glucagon-Secreting Cells ,Hyperaminoacidemia ,Glucagon receptor ,Glucagon receptor family ,Signal Transduction - Abstract
Glucagon supports glucose homeostasis by stimulating hepatic gluconeogenesis, in part by promoting the uptake and conversion of amino acids into gluconeogenic precursors. Genetic disruption or pharmacologic inhibition of glucagon signaling results in elevated plasma amino acids and compensatory glucagon hypersecretion involving expansion of pancreatic α cell mass. Recent findings indicate that hyperaminoacidemia triggers pancreatic α cell proliferation via an mTOR-dependent pathway. We confirm and extend these findings by demonstrating that glucagon pathway blockade selectively increases expression of the sodium-coupled neutral amino acid transporter Slc38a5 in a subset of highly proliferative α cells and that Slc38a5 controls the pancreatic response to glucagon pathway blockade; most notably, mice deficient in Slc38a5 exhibit markedly decreased α cell hyperplasia to glucagon pathway blockade-induced hyperaminoacidemia. These results show that Slc38a5 is a key component of the feedback circuit between glucagon receptor signaling in the liver and amino-acid-dependent regulation of pancreatic α cell mass in mice.
- Published
- 2017
48. Activin A more prominently regulates muscle mass in primates than does GDF8
- Author
-
Jason Mastaitis, Andrew J. Murphy, Jesus A. Trejos, Stephen Jaspers, Jeffrey Pangilinan, Trevor Stitt, Erqian Na, Esther Latres, Yu Bai, George D. Yancopoulos, Lawrence Miloscio, Jesper Gromada, Ashique Rafique, Wen Fury, Jee Hae Kim, Tobias Willer, Katie Cavino, Haruka Okamoto, and Angelos Papatheodorou
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,animal structures ,Science ,Activin Receptors, Type II ,Regulator ,General Physics and Astronomy ,Mice, SCID ,Biology ,Muscle mass ,Dexamethasone ,Article ,General Biochemistry, Genetics and Molecular Biology ,Body Mass Index ,Muscle hypertrophy ,Mice ,03 medical and health sciences ,Isometric Contraction ,Internal medicine ,medicine ,Animals ,Humans ,Muscle, Skeletal ,Multidisciplinary ,Antibodies, Monoclonal ,Hypertrophy ,General Chemistry ,Activin receptor ,Myostatin ,Activins ,Rats ,Blockade ,Mice, Inbred C57BL ,Activin a ,Macaca fascicularis ,030104 developmental biology ,Endocrinology ,embryonic structures ,biology.protein ,Muscle Hypotonia ,Antibody ,hormones, hormone substitutes, and hormone antagonists ,Transforming growth factor - Abstract
Growth and differentiation factor 8 (GDF8) is a TGF-β superfamily member, and negative regulator of skeletal muscle mass. GDF8 inhibition results in prominent muscle growth in mice, but less impressive hypertrophy in primates, including man. Broad TGF-β inhibition suggests another family member negatively regulates muscle mass, and its blockade enhances muscle growth seen with GDF8-specific inhibition. Here we show that activin A is the long-sought second negative muscle regulator. Activin A specific inhibition, on top of GDF8 inhibition, leads to pronounced muscle hypertrophy and force production in mice and monkeys. Inhibition of these two ligands mimics the hypertrophy seen with broad TGF-β blockers, while avoiding the adverse effects due to inhibition of multiple family members. Altogether, we identify activin A as a second negative regulator of muscle mass, and suggest that inhibition of both ligands provides a preferred therapeutic approach, which maximizes the benefit:risk ratio for muscle diseases in man., Inhibition of GDF8 increases muscle mass in mice, but is less effective in monkeys and humans. Here the authors show that activin A also inhibits muscle hypertrophy and that concomitant inhibition of activin A and GDF8 synergistically increases muscle mass in mice and non-human primates.
- Published
- 2017
49. ANGPTL8 requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clearance[S]
- Author
-
Helen H. Hobbs, Corey A. Alexa-Braun, Lisa M. Shihanian, Jesper Gromada, Buckler David R, Jonathan C. Cohen, Sandra Kleiner, Viktoria Gusarova, George D. Yancopoulos, Panayiotis Stevis, Andrew J. Murphy, Ivory J. Mintah, Serena Banfi, and Jorge F. Haller
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Peptide Hormones ,Amino Acid Motifs ,QD415-436 ,CHO Cells ,030204 cardiovascular system & hematology ,Biology ,Peptide hormone ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Cricetulus ,Angiopoietin-Like Protein 8 ,Internal medicine ,ANGPTL3 ,Cricetinae ,Blocking antibody ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Research Articles ,Triglycerides ,Angiopoietin-Like Protein 3 ,Hypertriglyceridemia ,angiopoietin-like 8 ,Lipoprotein lipase ,angiopoietin-like 3 ,Chinese hamster ovary cell ,HEK 293 cells ,digestive, oral, and skin physiology ,nutritional and metabolic diseases ,blocking antibody ,Cell Biology ,biology.organism_classification ,Lipoprotein Lipase ,030104 developmental biology ,Secretory protein ,Angiopoietin-like Proteins ,HEK293 Cells ,lipids (amino acids, peptides, and proteins) - Abstract
Angiopoietin-like (ANGPTL)3 and ANGPTL8 are secreted proteins and inhibitors of LPL-mediated plasma triglyceride (TG) clearance. It is unclear how these two ANGPTL proteins interact to regulate LPL activity. ANGPTL3 inhibits LPL activity and increases serum TG independent of ANGPTL8. These effects are reversed with an ANGPTL3 blocking antibody. Here, we show that ANGPTL8, although it possesses a functional inhibitory motif, is inactive by itself and requires ANGPTL3 expression to inhibit LPL and increase plasma TG. Using a mutated form of ANGPTL3 that lacks LPL inhibitory activity, we demonstrate that ANGPTL3 activity is not required for its ability to activate ANGPTL8. Moreover, coexpression of ANGPTL3 and ANGPTL8 leads to a far more efficacious increase in TG in mice than ANGPTL3 alone, suggesting the major inhibitory activity of this complex derives from ANGPTL8. An antibody to the C terminus of ANGPTL8 reversed LPL inhibition by ANGPTL8 in the presence of ANGPTL3. The antibody did not disrupt the ANGPTL8:ANGPTL3 complex, but came in close proximity to the LPL inhibitory motif in the N terminus of ANGPTL8. Collectively, these data show that ANGPTL8 has a functional LPL inhibitory motif, but only inhibits LPL and increases plasma TG levels in mice in the presence of ANGPTL3.
- Published
- 2017
50. Inhibition of PCSK9 does not improve lipopolysaccharide-induced mortality in mice
- Author
-
Angel Loza Valdes, Norma N. Anderson, J.-M. Berger, Jay D. Horton, and Jesper Gromada
- Subjects
0301 basic medicine ,Genetically modified mouse ,Lipopolysaccharides ,medicine.medical_specialty ,Lipopolysaccharide ,Inflammation ,QD415-436 ,Antibodies, Monoclonal, Humanized ,Biochemistry ,LDL ,Sepsis ,Cohort Studies ,PCSK9 ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Endocrinology ,Internal medicine ,medicine ,Animals ,Humans ,Protease Inhibitors ,Research Articles ,Cholesterol ,business.industry ,endotoxemia ,PCSK9 Inhibitors ,lipopolysaccharide ,Antibodies, Monoclonal ,cholesterol ,Cell Biology ,Cholesterol, LDL ,medicine.disease ,Survival Analysis ,3. Good health ,030104 developmental biology ,chemistry ,LDL receptor ,Knockout mouse ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,business - Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that targets LDL receptors (LDLRs) for degradation in liver. Blocking the interaction of PCSK9 with the LDLR potently reduces plasma LDL cholesterol levels and cardiovascular events. Recently, it has been suggested that inhibition of PCSK9 might also improve outcomes in mice and humans with sepsis, possibly by increasing LDLR-mediated clearance of endotoxins. Sepsis is a complication of a severe microbial infection that has shared pathways with lipid metabolism. Here, we tested whether anti-PCSK9 antibodies prevent death from lipopolysaccharide (LPS)-induced endotoxemia. Mice were administered PCSK9 antibodies prior to, or shortly after, injecting LPS. In both scenarios, the administration of PCSK9 antibodies did not alter endotoxemia-induced mortality. Afterward, we determined whether the complete absence of PCSK9 improved endotoxemia-induced mortality in mice with the germ-line deletion of Pcsk9. Similarly, PCSK9 knockout mice were not protected from LPS-induced death. To determine whether low LDLR expression increased LPS-induced mortality, Ldlr−/− mice and PCSK9 transgenic mice were studied after injection of LPS. Endotoxemia-induced mortality was not altered in either mouse model. In a human cohort, we observed no correlation between plasma inflammation markers with total cholesterol levels, LDL cholesterol, and PCSK9. Combined, our data demonstrate that PCSK9 inhibition provides no protection from LPS-induced mortality in mice.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.