Skip to main content
Top
Published in: Diabetologia 7/2008

01-07-2008 | Review

Newly identified loci highlight beta cell dysfunction as a key cause of type 2 diabetes: Where are the insulin resistance genes?

Author: J. C. Florez

Published in: Diabetologia | Issue 7/2008

Login to get access

Abstract

Although type 2 diabetes has been traditionally understood as a metabolic disorder initiated by insulin resistance, it has recently become apparent that an impairment in insulin secretion contributes to its manifestation and may play a prominent role in its early pathophysiology. The genetic dissection of Mendelian and, more recently, polygenic types of diabetes confirms the notion that primary defects in insulin synthesis, processing and/or secretion often give rise to the common form of this disorder. This concept, first advanced with the discovery and physiological characterisation of various genetic subtypes of MODY, has been extended to other forms of monogenic diabetes (e.g. neonatal diabetes). It has also led to the identification of common risk variants via candidate gene approaches (e.g. the E23K polymorphism in KCNJ11 or common variants in the MODY genes), and it has been validated by the description of the robust physiological effects conferred by polymorphisms in the TCF7L2 gene. More recently, the completion and integration of genome-wide association scans for this disease has uncovered a number of heretofore unsuspected variants, several of which also affect insulin secretion. This review provides an up-to-date account of genetic loci that influence risk of common type 2 diabetes via impairment of beta cell function, outlines their presumed mechanisms of action, and places them in the context of gene–gene and/or gene–environment interactions. Finally, a strategy for the analogous discovery of insulin resistance genes is proposed.
Literature
1.
go back to reference Yalow RS, Berson SA (1960) Immunoassay of endogenous plasma insulin in man. J Clin Invest 39:1157–1175PubMedCrossRef Yalow RS, Berson SA (1960) Immunoassay of endogenous plasma insulin in man. J Clin Invest 39:1157–1175PubMedCrossRef
2.
go back to reference Reaven GM, Bernstein R, Davis B, Olefsky JM (1976) Nonketotic diabetes mellitus: insulin deficiency or insulin resistance? Am J Med 60:80–88PubMedCrossRef Reaven GM, Bernstein R, Davis B, Olefsky JM (1976) Nonketotic diabetes mellitus: insulin deficiency or insulin resistance? Am J Med 60:80–88PubMedCrossRef
3.
go back to reference Ferrannini E (1998) Insulin resistance versus insulin deficiency in non-insulin-dependent diabetes mellitus: problems and prospects. Endocr Rev 19:477–490PubMedCrossRef Ferrannini E (1998) Insulin resistance versus insulin deficiency in non-insulin-dependent diabetes mellitus: problems and prospects. Endocr Rev 19:477–490PubMedCrossRef
4.
go back to reference Kahn SE (2003) The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46:3–19PubMedCrossRef Kahn SE (2003) The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46:3–19PubMedCrossRef
5.
go back to reference Lillioja S, Mott DM, Spraul M et al (1993) Insulin resistance and insulin secretory dysfunction as precursors of non-insulin-dependent diabetes mellitus. Prospective studies of Pima Indians. N Engl J Med 329:1988–1992PubMedCrossRef Lillioja S, Mott DM, Spraul M et al (1993) Insulin resistance and insulin secretory dysfunction as precursors of non-insulin-dependent diabetes mellitus. Prospective studies of Pima Indians. N Engl J Med 329:1988–1992PubMedCrossRef
6.
go back to reference Haffner SM, Miettinen H, Gaskill SP, Stern MP (1995) Decreased insulin secretion and increased insulin resistance are independently related to the 7-year risk of NIDDM in Mexican-Americans. Diabetes 44:1386–1391PubMedCrossRef Haffner SM, Miettinen H, Gaskill SP, Stern MP (1995) Decreased insulin secretion and increased insulin resistance are independently related to the 7-year risk of NIDDM in Mexican-Americans. Diabetes 44:1386–1391PubMedCrossRef
7.
go back to reference Gerich JE (2000) Insulin resistance is not necessarily an essential component of type 2 diabetes. J Clin Endocrinol Metab 85:2113–2115PubMedCrossRef Gerich JE (2000) Insulin resistance is not necessarily an essential component of type 2 diabetes. J Clin Endocrinol Metab 85:2113–2115PubMedCrossRef
8.
go back to reference Pimenta W, Korytkowski M, Mitrakou A et al (1995) Pancreatic beta-cell dysfunction as the primary genetic lesion in NIDDM. Evidence from studies in normal glucose-tolerant individuals with a first-degree NIDDM relative. JAMA 273:1855–1861PubMedCrossRef Pimenta W, Korytkowski M, Mitrakou A et al (1995) Pancreatic beta-cell dysfunction as the primary genetic lesion in NIDDM. Evidence from studies in normal glucose-tolerant individuals with a first-degree NIDDM relative. JAMA 273:1855–1861PubMedCrossRef
9.
go back to reference Polonsky KS, Sturis J, Bell GI (1996) Non-insulin-dependent diabetes mellitus—a genetically programmed failure of the beta cell to compensate for insulin resistance. N Engl J Med 334:777–783PubMedCrossRef Polonsky KS, Sturis J, Bell GI (1996) Non-insulin-dependent diabetes mellitus—a genetically programmed failure of the beta cell to compensate for insulin resistance. N Engl J Med 334:777–783PubMedCrossRef
10.
go back to reference Fajans SS, Bell GI, Polonsky KS (2001) Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 345:971–980PubMedCrossRef Fajans SS, Bell GI, Polonsky KS (2001) Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 345:971–980PubMedCrossRef
11.
go back to reference Vaxillaire M, Froguel P (2006) Genetic basis of maturity-onset diabetes of the young. Endocrinol Metab Clin N Am 35:371–384CrossRef Vaxillaire M, Froguel P (2006) Genetic basis of maturity-onset diabetes of the young. Endocrinol Metab Clin N Am 35:371–384CrossRef
12.
go back to reference Bell G, Polonsky KS (2001) Diabetes mellitus and genetically programmed defects in beta-cell function. Nature 414:788–791PubMedCrossRef Bell G, Polonsky KS (2001) Diabetes mellitus and genetically programmed defects in beta-cell function. Nature 414:788–791PubMedCrossRef
13.
go back to reference Florez JC, Hirschhorn JN, Altshuler D (2003) The inherited basis of diabetes mellitus: implications for the genetic analysis of complex traits. Annu Rev Genom Hum Genet 4:257–291CrossRef Florez JC, Hirschhorn JN, Altshuler D (2003) The inherited basis of diabetes mellitus: implications for the genetic analysis of complex traits. Annu Rev Genom Hum Genet 4:257–291CrossRef
14.
go back to reference Love-Gregory LD, Wasson J, Ma J et al (2004) A common polymorphism in the upstream promoter region of the hepatocyte nuclear factor-4α gene on chromosome 20 q is associated with type 2 diabetes and appears to contribute to the evidence for linkage in an Ashkenazi Jewish population. Diabetes 53:1134–1140PubMedCrossRef Love-Gregory LD, Wasson J, Ma J et al (2004) A common polymorphism in the upstream promoter region of the hepatocyte nuclear factor-4α gene on chromosome 20 q is associated with type 2 diabetes and appears to contribute to the evidence for linkage in an Ashkenazi Jewish population. Diabetes 53:1134–1140PubMedCrossRef
15.
go back to reference Silander K, Mohlke KL, Scott LJ et al (2004) Genetic variation near the hepatocyte nuclear factor-4α gene predicts susceptibility to type 2 diabetes. Diabetes 53:1141–1149PubMedCrossRef Silander K, Mohlke KL, Scott LJ et al (2004) Genetic variation near the hepatocyte nuclear factor-4α gene predicts susceptibility to type 2 diabetes. Diabetes 53:1141–1149PubMedCrossRef
16.
go back to reference Weedon MN, Owen KR, Shields B et al (2004) Common variants of the hepatocyte nuclear factor-4α P2 promoter are associated with type 2 diabetes in the U.K. population. Diabetes 53:3002–3006PubMedCrossRef Weedon MN, Owen KR, Shields B et al (2004) Common variants of the hepatocyte nuclear factor-4α P2 promoter are associated with type 2 diabetes in the U.K. population. Diabetes 53:3002–3006PubMedCrossRef
17.
go back to reference Hansen SK, Rose CS, Glumer C et al (2005) Variation near the hepatocyte nuclear factor (HNF)-4α gene associates with type 2 diabetes in the Danish population. Diabetologia 48:452–458PubMedCrossRef Hansen SK, Rose CS, Glumer C et al (2005) Variation near the hepatocyte nuclear factor (HNF)-4α gene associates with type 2 diabetes in the Danish population. Diabetologia 48:452–458PubMedCrossRef
18.
go back to reference Winckler W, Graham RR, de Bakker PIW et al (2005) Association testing of variants in the hepatocyte nuclear factor 4α gene with risk of type 2 diabetes in 7,883 people. Diabetes 54:886–892PubMedCrossRef Winckler W, Graham RR, de Bakker PIW et al (2005) Association testing of variants in the hepatocyte nuclear factor 4α gene with risk of type 2 diabetes in 7,883 people. Diabetes 54:886–892PubMedCrossRef
19.
go back to reference Winckler W, Burtt NP, Holmkvist J et al (2005) Association of common variation in the HNF1α gene region with risk of type 2 diabetes. Diabetes 54:2336–2342PubMedCrossRef Winckler W, Burtt NP, Holmkvist J et al (2005) Association of common variation in the HNF1α gene region with risk of type 2 diabetes. Diabetes 54:2336–2342PubMedCrossRef
20.
go back to reference Weedon MN, Owen KR, Shields B et al (2005) A large-scale association analysis of common variation of the HNF1α gene with type 2 diabetes in the U.K. Caucasian population. Diabetes 54:2487–2491PubMedCrossRef Weedon MN, Owen KR, Shields B et al (2005) A large-scale association analysis of common variation of the HNF1α gene with type 2 diabetes in the U.K. Caucasian population. Diabetes 54:2487–2491PubMedCrossRef
21.
go back to reference Bonnycastle LL, Willer CJ, Conneely KN et al (2006) Common variants in Maturity-Onset Diabetes of the Young genes contribute to risk of type 2 diabetes in Finns. Diabetes 55:2534–2540PubMedCrossRef Bonnycastle LL, Willer CJ, Conneely KN et al (2006) Common variants in Maturity-Onset Diabetes of the Young genes contribute to risk of type 2 diabetes in Finns. Diabetes 55:2534–2540PubMedCrossRef
22.
go back to reference Holmkvist J, Cervin C, Lyssenko V et al (2006) Common variants in HNF-1α and risk of type 2 diabetes. Diabetologia 49:2882–2891PubMedCrossRef Holmkvist J, Cervin C, Lyssenko V et al (2006) Common variants in HNF-1α and risk of type 2 diabetes. Diabetologia 49:2882–2891PubMedCrossRef
23.
go back to reference Willer CJ, Bonnycastle LL, Conneely KN et al (2007) Screening of 134 single nucleotide polymorphisms (SNPs) previously associated with type 2 diabetes replicates association with 12 SNPs in nine genes. Diabetes 56:256–264PubMedCrossRef Willer CJ, Bonnycastle LL, Conneely KN et al (2007) Screening of 134 single nucleotide polymorphisms (SNPs) previously associated with type 2 diabetes replicates association with 12 SNPs in nine genes. Diabetes 56:256–264PubMedCrossRef
25.
go back to reference Holmkvist J, Almgren P, Lyssenko V et al (2008) Common variants in MODY genes and future risk of type 2 diabetes. Diabetes (in press). DOI 10.2337/db06–1464 Holmkvist J, Almgren P, Lyssenko V et al (2008) Common variants in MODY genes and future risk of type 2 diabetes. Diabetes (in press). DOI 10.​2337/​db06–1464
26.
go back to reference Winckler W, Weedon MN, Graham RR et al (2007) Evaluation of common variants in the six known Maturity-Onset Diabetes of the Young (MODY) genes for association with type 2 diabetes. Diabetes 56:685–693PubMedCrossRef Winckler W, Weedon MN, Graham RR et al (2007) Evaluation of common variants in the six known Maturity-Onset Diabetes of the Young (MODY) genes for association with type 2 diabetes. Diabetes 56:685–693PubMedCrossRef
27.
go back to reference Gudmundsson J, Sulem P, Steinthorsdottir V et al (2007) Two variants on chromosome 17 confer prostate cancer risk, and the one in TCF2 protects against type 2 diabetes. Nat Genet 39:977–983PubMedCrossRef Gudmundsson J, Sulem P, Steinthorsdottir V et al (2007) Two variants on chromosome 17 confer prostate cancer risk, and the one in TCF2 protects against type 2 diabetes. Nat Genet 39:977–983PubMedCrossRef
28.
go back to reference Weedon MN, Clark VJ, Qian Y et al (2006) A common haplotype of the glucokinase gene alters fasting glucose and birth weight: association in six studies and population-genetics analyses. Am J Hum Genet 79:991–1001PubMedCrossRef Weedon MN, Clark VJ, Qian Y et al (2006) A common haplotype of the glucokinase gene alters fasting glucose and birth weight: association in six studies and population-genetics analyses. Am J Hum Genet 79:991–1001PubMedCrossRef
29.
go back to reference Kavvoura FK, Ioannidis JP (2005) Ala45Thr polymorphism of the NEUROD1 gene and diabetes susceptibility: a meta-analysis. Hum Genet 116:192–199PubMedCrossRef Kavvoura FK, Ioannidis JP (2005) Ala45Thr polymorphism of the NEUROD1 gene and diabetes susceptibility: a meta-analysis. Hum Genet 116:192–199PubMedCrossRef
30.
go back to reference Inoue H, Tanizawa Y, Wasson J et al (1998) A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome). Nat Genet 20:143–148PubMedCrossRef Inoue H, Tanizawa Y, Wasson J et al (1998) A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome). Nat Genet 20:143–148PubMedCrossRef
31.
go back to reference Hardy C, Khanim F, Torres R et al (1999) Clinical and molecular genetic analysis of 19 Wolfram syndrome kindreds demonstrating a wide spectrum of mutations in WFS1. Am J Hum Genet 65:1279–1290PubMedCrossRef Hardy C, Khanim F, Torres R et al (1999) Clinical and molecular genetic analysis of 19 Wolfram syndrome kindreds demonstrating a wide spectrum of mutations in WFS1. Am J Hum Genet 65:1279–1290PubMedCrossRef
32.
go back to reference Sandhu MS, Weedon MN, Fawcett KA et al (2007) Common variants in WFS1 confer risk of type 2 diabetes. Nat Genet 39:951–953PubMedCrossRef Sandhu MS, Weedon MN, Fawcett KA et al (2007) Common variants in WFS1 confer risk of type 2 diabetes. Nat Genet 39:951–953PubMedCrossRef
33.
go back to reference Franks PW, Rolandsson O, Debenham SL et al (2008) Replication of the association between variants in WFS1 and risk of type 2 diabetes in European populations. Diabetologia 51:458–463PubMedCrossRef Franks PW, Rolandsson O, Debenham SL et al (2008) Replication of the association between variants in WFS1 and risk of type 2 diabetes in European populations. Diabetologia 51:458–463PubMedCrossRef
34.
go back to reference Florez JC, Jablonski KA, McAteer J et al (2008) Testing of diabetes-associated WFS1 polymorphisms in the Diabetes Prevention Program. Diabetologia 51:451–457PubMedCrossRef Florez JC, Jablonski KA, McAteer J et al (2008) Testing of diabetes-associated WFS1 polymorphisms in the Diabetes Prevention Program. Diabetologia 51:451–457PubMedCrossRef
35.
go back to reference Aguilar-Bryan L, Bryan J (1999) Molecular biology of adenosine triphosphate-sensitive potassium channels. Endocr Rev 20:101–135PubMedCrossRef Aguilar-Bryan L, Bryan J (1999) Molecular biology of adenosine triphosphate-sensitive potassium channels. Endocr Rev 20:101–135PubMedCrossRef
36.
go back to reference Ashcroft FM, Gribble FM (1999) ATP-sensitive K+ channels and insulin secretion: their role in health and disease. Diabetologia 42:903–919PubMedCrossRef Ashcroft FM, Gribble FM (1999) ATP-sensitive K+ channels and insulin secretion: their role in health and disease. Diabetologia 42:903–919PubMedCrossRef
37.
go back to reference Altshuler D, Hirschhorn JN, Klannemark M et al (2000) The common PPARγ Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet 26:76–80PubMedCrossRef Altshuler D, Hirschhorn JN, Klannemark M et al (2000) The common PPARγ Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet 26:76–80PubMedCrossRef
38.
go back to reference Sakura H, Wat N, Horton V, Millns H, Turner RC, Ashcroft FM (1996) Sequence variations in the human Kir6.2 gene, a subunit of the beta-cell ATP-sensitive K-channel: no association with NIDDM in white Caucasian subjects or evidence of abnormal function when expressed in vitro. Diabetologia 39:1233–1236PubMedCrossRef Sakura H, Wat N, Horton V, Millns H, Turner RC, Ashcroft FM (1996) Sequence variations in the human Kir6.2 gene, a subunit of the beta-cell ATP-sensitive K-channel: no association with NIDDM in white Caucasian subjects or evidence of abnormal function when expressed in vitro. Diabetologia 39:1233–1236PubMedCrossRef
39.
go back to reference Inoue H, Ferrer J, Warren-Perry M et al (1997) Sequence variants in the pancreatic islet beta-cell inwardly rectifying K+ channel Kir6.2 (Bir) gene: identification and lack of role in Caucasian patients with NIDDM. Diabetes 46:502–507PubMedCrossRef Inoue H, Ferrer J, Warren-Perry M et al (1997) Sequence variants in the pancreatic islet beta-cell inwardly rectifying K+ channel Kir6.2 (Bir) gene: identification and lack of role in Caucasian patients with NIDDM. Diabetes 46:502–507PubMedCrossRef
40.
go back to reference Hansen L, Echwald SM, Hansen T, Urhammer SA, Clausen JO, Pedersen O (1997) Amino acid polymorphisms in the ATP-regulatable inward rectifier Kir6.2 and their relationships to glucose- and tolbutamide-induced insulin secretion, the insulin sensitivity index, and NIDDM. Diabetes 46:508–512PubMedCrossRef Hansen L, Echwald SM, Hansen T, Urhammer SA, Clausen JO, Pedersen O (1997) Amino acid polymorphisms in the ATP-regulatable inward rectifier Kir6.2 and their relationships to glucose- and tolbutamide-induced insulin secretion, the insulin sensitivity index, and NIDDM. Diabetes 46:508–512PubMedCrossRef
41.
go back to reference Hani EH, Boutin P, Durand E et al (1998) Missense mutations in the pancreatic islet beta cell inwardly rectifying K+ channel gene (KIR6.2/BIR): a meta-analysis suggests a role in the polygenic basis of type II diabetes mellitus in Caucasians. Diabetologia 41:1511–1515PubMedCrossRef Hani EH, Boutin P, Durand E et al (1998) Missense mutations in the pancreatic islet beta cell inwardly rectifying K+ channel gene (KIR6.2/BIR): a meta-analysis suggests a role in the polygenic basis of type II diabetes mellitus in Caucasians. Diabetologia 41:1511–1515PubMedCrossRef
42.
go back to reference Gloyn AL, Hashim Y, Ashcroft SJ, Ashfield R, Wiltshire S, Turner RC (2001) Association studies of variants in promoter and coding regions of beta-cell ATP-sensitive K-channel genes SUR1 and Kir6.2 with type 2 diabetes mellitus (UKPDS 53). Diabet Med 18:206–212PubMedCrossRef Gloyn AL, Hashim Y, Ashcroft SJ, Ashfield R, Wiltshire S, Turner RC (2001) Association studies of variants in promoter and coding regions of beta-cell ATP-sensitive K-channel genes SUR1 and Kir6.2 with type 2 diabetes mellitus (UKPDS 53). Diabet Med 18:206–212PubMedCrossRef
43.
go back to reference Gloyn AL, Weedon MN, Owen KR et al (2003) Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes 52:568–572PubMedCrossRef Gloyn AL, Weedon MN, Owen KR et al (2003) Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes 52:568–572PubMedCrossRef
44.
go back to reference Nielsen E-MD, Hansen L, Carstensen B et al (2003) The E23K variant of Kir6.2 associates with impaired post-OGTT serum insulin response and increased risk of type 2 diabetes. Diabetes 52:573–577PubMedCrossRef Nielsen E-MD, Hansen L, Carstensen B et al (2003) The E23K variant of Kir6.2 associates with impaired post-OGTT serum insulin response and increased risk of type 2 diabetes. Diabetes 52:573–577PubMedCrossRef
45.
go back to reference Love-Gregory L, Wasson J, Lin J, Skolnick G, Suarez B, Permutt MA (2003) E23K single nucleotide polymorphism in the islet ATP-sensitive potassium channel gene (Kir6.2) contributes as much to the risk of type II diabetes in Caucasians as the PPARγ Pro12Ala variant. Diabetologia 46:136–137PubMed Love-Gregory L, Wasson J, Lin J, Skolnick G, Suarez B, Permutt MA (2003) E23K single nucleotide polymorphism in the islet ATP-sensitive potassium channel gene (Kir6.2) contributes as much to the risk of type II diabetes in Caucasians as the PPARγ Pro12Ala variant. Diabetologia 46:136–137PubMed
46.
go back to reference Florez JC, Burtt N, de Bakker PIW et al (2004) Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53:1360–1368PubMedCrossRef Florez JC, Burtt N, de Bakker PIW et al (2004) Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53:1360–1368PubMedCrossRef
47.
go back to reference van Dam RM, Hoebee B, Seidell JC, Schaap MM, de Bruin TWA, Feskens EJM (2005) Common variants in the ATP-sensitive K+ channel genes KCNJ11 (Kir6.2) and ABCC8 (SUR1) in relation to glucose intolerance: population-based studies and meta-analyses. Diabet Med 22:590–598PubMedCrossRef van Dam RM, Hoebee B, Seidell JC, Schaap MM, de Bruin TWA, Feskens EJM (2005) Common variants in the ATP-sensitive K+ channel genes KCNJ11 (Kir6.2) and ABCC8 (SUR1) in relation to glucose intolerance: population-based studies and meta-analyses. Diabet Med 22:590–598PubMedCrossRef
48.
go back to reference Schwanstecher C, Meyer U, Schwanstecher M (2002) KIR6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic β-cell ATP-sensitive K+ channels. Diabetes 51:875–879PubMedCrossRef Schwanstecher C, Meyer U, Schwanstecher M (2002) KIR6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic β-cell ATP-sensitive K+ channels. Diabetes 51:875–879PubMedCrossRef
49.
go back to reference Riedel MJ, Boora P, Steckley D, de Vries G, Light PE (2003) Kir6.2 polymorphisms sensitize β-cell ATP-sensitive potassium channels to activation by acyl CoAs: A possible cellular mechanism for increased susceptibility to type 2 diabetes? Diabetes 52:2630–2635PubMedCrossRef Riedel MJ, Boora P, Steckley D, de Vries G, Light PE (2003) Kir6.2 polymorphisms sensitize β-cell ATP-sensitive potassium channels to activation by acyl CoAs: A possible cellular mechanism for increased susceptibility to type 2 diabetes? Diabetes 52:2630–2635PubMedCrossRef
50.
go back to reference Riedel MJ, Light PE (2005) Saturated and cis/trans unsaturated acyl CoA esters differentially regulate wild-type and polymorphic β-cell ATP-sensitive K+ channels. Diabetes 54:2070–2079PubMedCrossRef Riedel MJ, Light PE (2005) Saturated and cis/trans unsaturated acyl CoA esters differentially regulate wild-type and polymorphic β-cell ATP-sensitive K+ channels. Diabetes 54:2070–2079PubMedCrossRef
51.
go back to reference Gloyn AL, Pearson ER, Antcliff JF et al (2004) Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 350:1838–1849PubMedCrossRef Gloyn AL, Pearson ER, Antcliff JF et al (2004) Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 350:1838–1849PubMedCrossRef
52.
go back to reference Zeggini E, McCarthy MI (2007) TCF7L2: the biggest story in diabetes genetics since HLA? Diabetologia 50:1–4PubMedCrossRef Zeggini E, McCarthy MI (2007) TCF7L2: the biggest story in diabetes genetics since HLA? Diabetologia 50:1–4PubMedCrossRef
53.
go back to reference Grant SFA, Thorleifsson G, Reynisdottir I et al (2006) Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38:320–323PubMedCrossRef Grant SFA, Thorleifsson G, Reynisdottir I et al (2006) Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38:320–323PubMedCrossRef
54.
go back to reference Cauchi S, El Achhab Y, Choquet H et al (2007) TCF7L2 is reproducibly associated with type 2 diabetes in various ethnic groups: a global meta-analysis. J Mol Med 85:777–782PubMedCrossRef Cauchi S, El Achhab Y, Choquet H et al (2007) TCF7L2 is reproducibly associated with type 2 diabetes in various ethnic groups: a global meta-analysis. J Mol Med 85:777–782PubMedCrossRef
55.
56.
go back to reference Miyake K, Horikawa Y, Hara K et al (2008) Association of TCF7L2 polymorphisms with susceptibility to type 2 diabetes in 4,087 Japanese subjects. J Hum Genet 53:174–180PubMedCrossRef Miyake K, Horikawa Y, Hara K et al (2008) Association of TCF7L2 polymorphisms with susceptibility to type 2 diabetes in 4,087 Japanese subjects. J Hum Genet 53:174–180PubMedCrossRef
57.
go back to reference Chang YC, Chang TJ, Jiang YD et al (2007) Association study of the genetic polymorphisms of the transcription factor 7-like 2 (TCF7L2) gene and type 2 diabetes in the Chinese population. Diabetes 56:2631–2637PubMedCrossRef Chang YC, Chang TJ, Jiang YD et al (2007) Association study of the genetic polymorphisms of the transcription factor 7-like 2 (TCF7L2) gene and type 2 diabetes in the Chinese population. Diabetes 56:2631–2637PubMedCrossRef
58.
go back to reference Korinek V, Barker N, Moerer P et al (1998) Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet 19:379–383PubMedCrossRef Korinek V, Barker N, Moerer P et al (1998) Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet 19:379–383PubMedCrossRef
59.
60.
go back to reference Yi F, Brubaker PL, Jin T (2005) TCF-4 mediates cell type-specific regulation of proglucagon gene expression by β-catenin and glycogen synthase kinase-3β. J Biol Chem 280:1457–1464PubMedCrossRef Yi F, Brubaker PL, Jin T (2005) TCF-4 mediates cell type-specific regulation of proglucagon gene expression by β-catenin and glycogen synthase kinase-3β. J Biol Chem 280:1457–1464PubMedCrossRef
61.
go back to reference Florez JC, Jablonski KA, Bayley N et al (2006) TCF7L2 polymorphisms and progression to diabetes in the Diabetes Prevention Program. N Engl J Med 355:241–250PubMedCrossRef Florez JC, Jablonski KA, Bayley N et al (2006) TCF7L2 polymorphisms and progression to diabetes in the Diabetes Prevention Program. N Engl J Med 355:241–250PubMedCrossRef
62.
go back to reference Saxena R, Gianniny L, Burtt NP et al (2006) Common single nucleotide polymorphisms in TCF7L2 are reproducibly associated with type 2 diabetes and reduce the insulin response to glucose in nondiabetic individuals. Diabetes 55:2890–2895PubMedCrossRef Saxena R, Gianniny L, Burtt NP et al (2006) Common single nucleotide polymorphisms in TCF7L2 are reproducibly associated with type 2 diabetes and reduce the insulin response to glucose in nondiabetic individuals. Diabetes 55:2890–2895PubMedCrossRef
63.
go back to reference Damcott CM, Pollin TI, Reinhart LJ et al (2006) Polymorphisms in the transcription factor 7-like 2 (TCF7L2) gene are associated with type 2 diabetes in the Amish: Replication and evidence for a role in both insulin secretion and insulin resistance. Diabetes 55:2654–2659PubMedCrossRef Damcott CM, Pollin TI, Reinhart LJ et al (2006) Polymorphisms in the transcription factor 7-like 2 (TCF7L2) gene are associated with type 2 diabetes in the Amish: Replication and evidence for a role in both insulin secretion and insulin resistance. Diabetes 55:2654–2659PubMedCrossRef
64.
go back to reference Munoz J, Lok KH, Gower BA et al (2006) Polymorphism in the transcription factor 7-like 2 (TCF7L2) gene is associated with reduced insulin secretion in nondiabetic women. Diabetes 55:3630–3634PubMedCrossRef Munoz J, Lok KH, Gower BA et al (2006) Polymorphism in the transcription factor 7-like 2 (TCF7L2) gene is associated with reduced insulin secretion in nondiabetic women. Diabetes 55:3630–3634PubMedCrossRef
65.
go back to reference Lyssenko V, Lupi R, Marchetti P et al (2007) Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 117:2155–2163PubMedCrossRef Lyssenko V, Lupi R, Marchetti P et al (2007) Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 117:2155–2163PubMedCrossRef
66.
go back to reference Palmer ND, Lehtinen AB, Langefeld CD et al (2008) Association of TCF7L2 gene polymorphisms with reduced acute insulin response in Hispanic Americans. J Clin Endocrinol Metab 93:304–309PubMedCrossRef Palmer ND, Lehtinen AB, Langefeld CD et al (2008) Association of TCF7L2 gene polymorphisms with reduced acute insulin response in Hispanic Americans. J Clin Endocrinol Metab 93:304–309PubMedCrossRef
67.
go back to reference Schafer SA, Tschritter O, Machicao F et al (2007) Impaired glucagon-like peptide-1-induced insulin secretion in carriers of transcription factor 7-like 2 (TCF7L2) gene polymorphisms. Diabetologia 50:2443–2450PubMedCrossRef Schafer SA, Tschritter O, Machicao F et al (2007) Impaired glucagon-like peptide-1-induced insulin secretion in carriers of transcription factor 7-like 2 (TCF7L2) gene polymorphisms. Diabetologia 50:2443–2450PubMedCrossRef
68.
go back to reference Liu Z, Habener JF (2008) Glucagon-like peptide-1 activation of TCF7L2-dependent Wnt signaling enhances pancreatic beta cell proliferation. J Biol Chem 283:8723–8735PubMedCrossRef Liu Z, Habener JF (2008) Glucagon-like peptide-1 activation of TCF7L2-dependent Wnt signaling enhances pancreatic beta cell proliferation. J Biol Chem 283:8723–8735PubMedCrossRef
69.
go back to reference Shu L, Sauter NS, Schulthess FT, Matveyenko AV, Oberholzer J, Maedler K (2008) Transcription factor 7-like 2 regulates β-cell survival and function in human pancreatic islets. Diabetes 57:645–653PubMedCrossRef Shu L, Sauter NS, Schulthess FT, Matveyenko AV, Oberholzer J, Maedler K (2008) Transcription factor 7-like 2 regulates β-cell survival and function in human pancreatic islets. Diabetes 57:645–653PubMedCrossRef
70.
go back to reference Loos RJF, Franks PW, Francis RW et al (2007) TCF7L2 polymorphisms modulate proinsulin levels and β-cell function in a British Europid population. Diabetes 56:1943–1947PubMedCrossRef Loos RJF, Franks PW, Francis RW et al (2007) TCF7L2 polymorphisms modulate proinsulin levels and β-cell function in a British Europid population. Diabetes 56:1943–1947PubMedCrossRef
72.
go back to reference Kirchhoff K, Machicao F, Haupt A et al (2008) Polymorphisms in the TCF7L2, CDKAL1 and SLC30A8 genes are associated with impaired proinsulin conversion. Diabetologia 51:597–601PubMedCrossRef Kirchhoff K, Machicao F, Haupt A et al (2008) Polymorphisms in the TCF7L2, CDKAL1 and SLC30A8 genes are associated with impaired proinsulin conversion. Diabetologia 51:597–601PubMedCrossRef
73.
go back to reference Wang J, Kuusisto J, Vanttinen M et al (2007) Variants of transcription factor 7-like 2 (TCF7L2) gene predict conversion to type 2 diabetes in the Finnish Diabetes Prevention Study and are associated with impaired glucose regulation and impaired insulin secretion. Diabetologia 50:1192–1200PubMedCrossRef Wang J, Kuusisto J, Vanttinen M et al (2007) Variants of transcription factor 7-like 2 (TCF7L2) gene predict conversion to type 2 diabetes in the Finnish Diabetes Prevention Study and are associated with impaired glucose regulation and impaired insulin secretion. Diabetologia 50:1192–1200PubMedCrossRef
74.
go back to reference Sladek R, Rocheleau G, Rung J et al (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445:828–830CrossRef Sladek R, Rocheleau G, Rung J et al (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445:828–830CrossRef
75.
go back to reference Diabetes Genetics Initiative of Broad Institute of Harvard and MIT, Lund University and Novartis Institutes for BioMedical Research (2007) Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316:1331–1336CrossRef Diabetes Genetics Initiative of Broad Institute of Harvard and MIT, Lund University and Novartis Institutes for BioMedical Research (2007) Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316:1331–1336CrossRef
76.
go back to reference Zeggini E, Weedon MN, Lindgren CM et al (2007) Replication of genome-wide association signals in U.K. samples reveals risk loci for type 2 diabetes. Science 316:1336–1341PubMedCrossRef Zeggini E, Weedon MN, Lindgren CM et al (2007) Replication of genome-wide association signals in U.K. samples reveals risk loci for type 2 diabetes. Science 316:1336–1341PubMedCrossRef
77.
go back to reference Scott LJ, Mohlke KL, Bonnycastle LL et al (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316:1341–1345PubMedCrossRef Scott LJ, Mohlke KL, Bonnycastle LL et al (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316:1341–1345PubMedCrossRef
78.
go back to reference Steinthorsdottir V, Thorleifsson G, Reynisdottir I et al (2007) A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet 39:770–775PubMedCrossRef Steinthorsdottir V, Thorleifsson G, Reynisdottir I et al (2007) A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet 39:770–775PubMedCrossRef
79.
go back to reference Frayling TM, Timpson NJ, Weedon MN et al (2007) A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316:889–894PubMedCrossRef Frayling TM, Timpson NJ, Weedon MN et al (2007) A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316:889–894PubMedCrossRef
80.
go back to reference Salonen JT, Uimari P, Aalto JM et al (2007) Type 2 diabetes whole-genome association study in four populations: the DiaGen consortium. Am J Hum Genet 81:338–345PubMedCrossRef Salonen JT, Uimari P, Aalto JM et al (2007) Type 2 diabetes whole-genome association study in four populations: the DiaGen consortium. Am J Hum Genet 81:338–345PubMedCrossRef
81.
go back to reference Hayes MG, Pluzhnikov A, Miyake K et al (2007) Identification of type 2 diabetes genes in Mexican Americans through genome-wide association studies. Diabetes 56:3033–3044PubMedCrossRef Hayes MG, Pluzhnikov A, Miyake K et al (2007) Identification of type 2 diabetes genes in Mexican Americans through genome-wide association studies. Diabetes 56:3033–3044PubMedCrossRef
82.
go back to reference Hanson RL, Bogardus C, Duggan D et al (2007) A search for variants associated with young-onset type 2 diabetes in American Indians in a 100K genotyping array. Diabetes 56:3045–3052PubMedCrossRef Hanson RL, Bogardus C, Duggan D et al (2007) A search for variants associated with young-onset type 2 diabetes in American Indians in a 100K genotyping array. Diabetes 56:3045–3052PubMedCrossRef
83.
go back to reference Rampersaud E, Damcott CM, Fu M et al (2007) Identification of novel candidate genes for type 2 diabetes from a genome-wide association scan in the Old Order Amish: Evidence for replication from diabetes-related quantitative traits and from independent populations. Diabetes 56:3053–3062PubMedCrossRef Rampersaud E, Damcott CM, Fu M et al (2007) Identification of novel candidate genes for type 2 diabetes from a genome-wide association scan in the Old Order Amish: Evidence for replication from diabetes-related quantitative traits and from independent populations. Diabetes 56:3053–3062PubMedCrossRef
84.
go back to reference Florez JC, Manning AK, Dupuis J et al (2007) A 100 K genome-wide association scan for diabetes and related traits in the Framingham Heart Study: replication and integration with other genome-wide datasets. Diabetes 56:3063–3074PubMedCrossRef Florez JC, Manning AK, Dupuis J et al (2007) A 100 K genome-wide association scan for diabetes and related traits in the Framingham Heart Study: replication and integration with other genome-wide datasets. Diabetes 56:3063–3074PubMedCrossRef
85.
go back to reference Pascoe L, Tura A, Patel SK et al (2007) Common variants of the novel type 2 diabetes genes, CDKAL1 and HHEX/IDE, are associated with decreased pancreatic β-cell function. Diabetes 56:3101–3104PubMedCrossRef Pascoe L, Tura A, Patel SK et al (2007) Common variants of the novel type 2 diabetes genes, CDKAL1 and HHEX/IDE, are associated with decreased pancreatic β-cell function. Diabetes 56:3101–3104PubMedCrossRef
86.
go back to reference Grarup N, Rose CS, Andersson EA et al (2007) Studies of association of variants near the HHEX, CDKN2A/B and IGF2BP2 genes with type 2 diabetes and impaired insulin release in 10,705 Danish subjects: validation and extension of genome-wide association studies. Diabetes 56:3105–3111PubMedCrossRef Grarup N, Rose CS, Andersson EA et al (2007) Studies of association of variants near the HHEX, CDKN2A/B and IGF2BP2 genes with type 2 diabetes and impaired insulin release in 10,705 Danish subjects: validation and extension of genome-wide association studies. Diabetes 56:3105–3111PubMedCrossRef
87.
go back to reference Staiger H, Machicao F, Stefan N et al (2007) Polymorphisms within novel risk loci for type 2 diabetes determine beta-cell function. PLoS ONE 2:e832PubMedCrossRef Staiger H, Machicao F, Stefan N et al (2007) Polymorphisms within novel risk loci for type 2 diabetes determine beta-cell function. PLoS ONE 2:e832PubMedCrossRef
88.
go back to reference Palmer ND, Goodarzi MO, Langefeld CD et al (2008) Quantitative trait analysis of T2D susceptibility loci identified from whole genome association studies in the IRAS Family Study. Diabetes 57:1093–1100PubMedCrossRef Palmer ND, Goodarzi MO, Langefeld CD et al (2008) Quantitative trait analysis of T2D susceptibility loci identified from whole genome association studies in the IRAS Family Study. Diabetes 57:1093–1100PubMedCrossRef
89.
go back to reference Taylor SI (1992) Lilly Lecture: molecular mechanisms of insulin resistance. Lessons from patients with mutations in the insulin-receptor gene. Diabetes 41:1473–1490PubMedCrossRef Taylor SI (1992) Lilly Lecture: molecular mechanisms of insulin resistance. Lessons from patients with mutations in the insulin-receptor gene. Diabetes 41:1473–1490PubMedCrossRef
90.
go back to reference Panhuysen CIM, Cupples LA, Wilson PWF, Herbert AG, Myers RH, Meigs JB (2003) A genome scan for loci linked to quantitative insulin traits in persons without diabetes: the Framingham Offspring Study. Diabetologia 46:579–587PubMed Panhuysen CIM, Cupples LA, Wilson PWF, Herbert AG, Myers RH, Meigs JB (2003) A genome scan for loci linked to quantitative insulin traits in persons without diabetes: the Framingham Offspring Study. Diabetologia 46:579–587PubMed
91.
go back to reference Lyssenko V, Almgren P, Anevski D et al (2005) Genetic prediction of future type 2 diabetes. PLoS Med 2:e345PubMedCrossRef Lyssenko V, Almgren P, Anevski D et al (2005) Genetic prediction of future type 2 diabetes. PLoS Med 2:e345PubMedCrossRef
92.
go back to reference Vanttinen M, Nuutila P, Pihlajamaki J et al (2005) The effect of the Ala12 allele of the peroxisome proliferator-activated receptor-γ2 gene on skeletal muscle glucose uptake depends on obesity: a positron emission tomography study. J Clin Endocrinol Metab 90:4249–4254PubMedCrossRef Vanttinen M, Nuutila P, Pihlajamaki J et al (2005) The effect of the Ala12 allele of the peroxisome proliferator-activated receptor-γ2 gene on skeletal muscle glucose uptake depends on obesity: a positron emission tomography study. J Clin Endocrinol Metab 90:4249–4254PubMedCrossRef
93.
go back to reference Tonjes A, Scholz M, Loeffler M, Stumvoll M (2006) Association of Pro12Ala polymorphism in peroxisome proliferator-activated receptor γ with pre-diabetic phenotypes: meta-analysis of 57 studies on nondiabetic individuals. Diabetes Care 29:2489–2497PubMedCrossRef Tonjes A, Scholz M, Loeffler M, Stumvoll M (2006) Association of Pro12Ala polymorphism in peroxisome proliferator-activated receptor γ with pre-diabetic phenotypes: meta-analysis of 57 studies on nondiabetic individuals. Diabetes Care 29:2489–2497PubMedCrossRef
94.
go back to reference Florez JC, Jablonski KA, Sun MW et al (2007) Effects of the type 2 diabetes-associated PPARG P12A polymorphism on progression to diabetes and response to troglitazone. J Clin Endocrinol Metab 92:1502–1509PubMedCrossRef Florez JC, Jablonski KA, Sun MW et al (2007) Effects of the type 2 diabetes-associated PPARG P12A polymorphism on progression to diabetes and response to troglitazone. J Clin Endocrinol Metab 92:1502–1509PubMedCrossRef
95.
go back to reference Ludovico O, Pellegrini F, Di Paola R et al (2007) Heterogeneous effect of peroxisome proliferator-activated receptor γ2 Ala12 variant on type 2 diabetes risk. Obesity 15:1076–1081PubMedCrossRef Ludovico O, Pellegrini F, Di Paola R et al (2007) Heterogeneous effect of peroxisome proliferator-activated receptor γ2 Ala12 variant on type 2 diabetes risk. Obesity 15:1076–1081PubMedCrossRef
96.
go back to reference Bochenski J, Placha G, Wanic K et al (2006) New polymorphism of ENPP1 (PC-1) is associated with increased risk of type 2 diabetes among obese individuals. Diabetes 55:2626–2630PubMedCrossRef Bochenski J, Placha G, Wanic K et al (2006) New polymorphism of ENPP1 (PC-1) is associated with increased risk of type 2 diabetes among obese individuals. Diabetes 55:2626–2630PubMedCrossRef
97.
go back to reference Chandalia M, Grundy S, Adams-Huet B, Abate N (2007) Ethnic differences in the frequency of ENPP1/PC1 121Q genetic variant in the Dallas Heart Study cohort. J Diabetes Complicat 21:143–148PubMedCrossRef Chandalia M, Grundy S, Adams-Huet B, Abate N (2007) Ethnic differences in the frequency of ENPP1/PC1 121Q genetic variant in the Dallas Heart Study cohort. J Diabetes Complicat 21:143–148PubMedCrossRef
98.
go back to reference McAteer JB, Prudente S, Bacci S et al (2007) The ENPP1 K121Q polymorphism is associated with type 2 diabetes in European populations: Evidence from an updated meta-analysis in 42,042 subjects. Diabetes 57:1125–1130PubMedCrossRef McAteer JB, Prudente S, Bacci S et al (2007) The ENPP1 K121Q polymorphism is associated with type 2 diabetes in European populations: Evidence from an updated meta-analysis in 42,042 subjects. Diabetes 57:1125–1130PubMedCrossRef
99.
go back to reference Bacci S, De Cosmo S, Prudente S, Trischitta V (2007) ENPP1 gene, insulin resistance and related clinical outcomes. Curr Opin Clin Nutr Metab Care 10:403–409PubMedCrossRef Bacci S, De Cosmo S, Prudente S, Trischitta V (2007) ENPP1 gene, insulin resistance and related clinical outcomes. Curr Opin Clin Nutr Metab Care 10:403–409PubMedCrossRef
100.
go back to reference Frayling TM (2007) Genome-wide association studies provide new insights into type 2 diabetes aetiology. Nat Rev Genet 8:657–662PubMedCrossRef Frayling TM (2007) Genome-wide association studies provide new insights into type 2 diabetes aetiology. Nat Rev Genet 8:657–662PubMedCrossRef
Metadata
Title
Newly identified loci highlight beta cell dysfunction as a key cause of type 2 diabetes: Where are the insulin resistance genes?
Author
J. C. Florez
Publication date
01-07-2008
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 7/2008
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-008-1025-9

Other articles of this Issue 7/2008

Diabetologia 7/2008 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.