Skip to main content
Top
Published in: Diabetologia 8/2011

01-08-2011 | Review

Regulation of hypoxia-inducible factor 1 and the loss of the cellular response to hypoxia in diabetes

Authors: C. F. Bento, P. Pereira

Published in: Diabetologia | Issue 8/2011

Login to get access

Abstract

Diabetes is frequently associated with hypoxia and is known to impair ischaemia-induced neovascularisation and other forms of adaptive cell and tissue responses to low oxygen levels. Hyperglycaemia appears to be the driving force of such deregulation. Recent data indicate that destabilisation of hypoxia-inducible factor 1 (HIF-1) is most likely the event that transduces hyperglycaemia into the loss of the cellular response to hypoxia in most diabetic complications. HIF-1 is a critical transcription factor involved in oxygen homeostasis that regulates a variety of adaptive responses to hypoxia, including angiogenesis, metabolic reprogramming and survival. Thus, destabilisation of HIF-1 is likely to have a negative impact on cell and tissue adaptation to low oxygen. Indeed, destabilisation of HIF-1 by high glucose levels has serious consequences in various organs and tissues, including myocardial collateralisation, wound healing, renal, neural and retinal function, as a result of poor cell and tissue responses to low oxygen. This review aims to integrate and summarise some of the most recent developments, including new proposed molecular models, on this research topic, particularly in terms of their implications for potential therapeutic approaches for the prevention or treatment of some of the diabetic complications characterised by impaired cellular and tissue responses to hypoxia.
Literature
1.
go back to reference Wang GL, Jiang BH, Rue EA, Semenza GL (1995) Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA 92:5510–5514PubMedCrossRef Wang GL, Jiang BH, Rue EA, Semenza GL (1995) Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA 92:5510–5514PubMedCrossRef
2.
go back to reference Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE (1991) Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene. Proc Natl Acad Sci USA 88:5680–5684PubMedCrossRef Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE (1991) Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene. Proc Natl Acad Sci USA 88:5680–5684PubMedCrossRef
3.
go back to reference Semenza GL, Wang GL (1992) A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12:5447–5454PubMed Semenza GL, Wang GL (1992) A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12:5447–5454PubMed
4.
go back to reference Huang LE, Gu J, Schau M, Bunn HF (1998) Regulation of hypoxia-inducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 95:7987–7992PubMedCrossRef Huang LE, Gu J, Schau M, Bunn HF (1998) Regulation of hypoxia-inducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 95:7987–7992PubMedCrossRef
5.
go back to reference Salceda S, Caro J (1997) Hypoxia-inducible factor 1α (HIF-1α) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes. J Biol Chem 272:22642–22647PubMedCrossRef Salceda S, Caro J (1997) Hypoxia-inducible factor 1α (HIF-1α) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes. J Biol Chem 272:22642–22647PubMedCrossRef
6.
go back to reference Huang LE, Arany Z, Livingston DM, Bunn HF (1996) Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its alpha subunit. J Biol Chem 271:32253–32259PubMedCrossRef Huang LE, Arany Z, Livingston DM, Bunn HF (1996) Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its alpha subunit. J Biol Chem 271:32253–32259PubMedCrossRef
7.
go back to reference Yu AY, Frid MG, Shimoda LA, Wiener CM, Stenmark K, Semenza GL (1998) Temporal, spatial, and oxygen-regulated expression of hypoxia-inducible factor-1 in the lung. Am J Physiol 275:L818–826PubMed Yu AY, Frid MG, Shimoda LA, Wiener CM, Stenmark K, Semenza GL (1998) Temporal, spatial, and oxygen-regulated expression of hypoxia-inducible factor-1 in the lung. Am J Physiol 275:L818–826PubMed
8.
go back to reference Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J (2003) HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1α in normoxia. EMBO J 22:4082–4090PubMedCrossRef Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J (2003) HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1α in normoxia. EMBO J 22:4082–4090PubMedCrossRef
9.
go back to reference Hon WC, Wilson MI, Harlos K et al (2002) Structural basis for the recognition of hydroxyproline in HIF-1α by pVHL. Nature 417:975–978PubMedCrossRef Hon WC, Wilson MI, Harlos K et al (2002) Structural basis for the recognition of hydroxyproline in HIF-1α by pVHL. Nature 417:975–978PubMedCrossRef
10.
go back to reference Bruick RK, McKnight SL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294:1337–1340PubMedCrossRef Bruick RK, McKnight SL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294:1337–1340PubMedCrossRef
11.
go back to reference Ivan M, Kondo K, Yang H et al (2001) HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292:464–468PubMedCrossRef Ivan M, Kondo K, Yang H et al (2001) HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292:464–468PubMedCrossRef
12.
go back to reference Jaakkola P, Mole DR, Tian YM et al (2001) Targeting of HIF-α to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292:468–472PubMedCrossRef Jaakkola P, Mole DR, Tian YM et al (2001) Targeting of HIF-α to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292:468–472PubMedCrossRef
13.
go back to reference Salnikow K, Donald SP, Bruick RK, Zhitkovich A, Phang JM, Kasprzak KS (2004) Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J Biol Chem 279:40337–40344PubMedCrossRef Salnikow K, Donald SP, Bruick RK, Zhitkovich A, Phang JM, Kasprzak KS (2004) Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J Biol Chem 279:40337–40344PubMedCrossRef
14.
go back to reference Wang GL, Semenza GL (1993) Desferrioxamine induces erythropoietin gene expression and hypoxia-inducible factor 1 DNA-binding activity: implications for models of hypoxia signal transduction. Blood 82:3610–3615PubMed Wang GL, Semenza GL (1993) Desferrioxamine induces erythropoietin gene expression and hypoxia-inducible factor 1 DNA-binding activity: implications for models of hypoxia signal transduction. Blood 82:3610–3615PubMed
15.
go back to reference Yuan Y, Hilliard G, Ferguson T, Millhorn DE (2003) Cobalt inhibits the interaction between hypoxia-inducible factor-α and von Hippel–Lindau protein by direct binding to hypoxia-inducible factor-α. J Biol Chem 278:15911–15916PubMedCrossRef Yuan Y, Hilliard G, Ferguson T, Millhorn DE (2003) Cobalt inhibits the interaction between hypoxia-inducible factor-α and von Hippel–Lindau protein by direct binding to hypoxia-inducible factor-α. J Biol Chem 278:15911–15916PubMedCrossRef
16.
go back to reference Hewitson KS, Lienard BM, McDonough MA et al (2007) Structural and mechanistic studies on the inhibition of the hypoxia-inducible transcription factor hydroxylases by tricarboxylic acid cycle intermediates. J Biol Chem 282:3293–3301PubMedCrossRef Hewitson KS, Lienard BM, McDonough MA et al (2007) Structural and mechanistic studies on the inhibition of the hypoxia-inducible transcription factor hydroxylases by tricarboxylic acid cycle intermediates. J Biol Chem 282:3293–3301PubMedCrossRef
17.
go back to reference Selak MA, Armour SM, MacKenzie ED et al (2005) Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell 7:77–85PubMedCrossRef Selak MA, Armour SM, MacKenzie ED et al (2005) Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell 7:77–85PubMedCrossRef
18.
go back to reference Maxwell PH, Wiesener MS, Chang GW et al (1999) The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399:271–275PubMedCrossRef Maxwell PH, Wiesener MS, Chang GW et al (1999) The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399:271–275PubMedCrossRef
19.
go back to reference Cockman ME, Masson N, Mole DR et al (2000) Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel–Lindau tumor suppressor protein. J Biol Chem 275:25733–25741PubMedCrossRef Cockman ME, Masson N, Mole DR et al (2000) Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel–Lindau tumor suppressor protein. J Biol Chem 275:25733–25741PubMedCrossRef
20.
go back to reference Kamura T, Sato S, Iwai K, Czyzyk-Krzeska M, Conaway RC, Conaway JW (2000) Activation of HIF1alpha ubiquitination by a reconstituted von Hippel–Lindau (VHL) tumor suppressor complex. Proc Natl Acad Sci USA 97:10430–10435PubMedCrossRef Kamura T, Sato S, Iwai K, Czyzyk-Krzeska M, Conaway RC, Conaway JW (2000) Activation of HIF1alpha ubiquitination by a reconstituted von Hippel–Lindau (VHL) tumor suppressor complex. Proc Natl Acad Sci USA 97:10430–10435PubMedCrossRef
21.
go back to reference Ohh M, Park CW, Ivan M et al (2000) Ubiquitination of hypoxia-inducible factor requires direct binding to the β-domain of the von Hippel–Lindau protein. Nat Cell Biol 2:423–427PubMedCrossRef Ohh M, Park CW, Ivan M et al (2000) Ubiquitination of hypoxia-inducible factor requires direct binding to the β-domain of the von Hippel–Lindau protein. Nat Cell Biol 2:423–427PubMedCrossRef
22.
go back to reference Iwai K, Yamanaka K, Kamura T et al (1999) Identification of the von Hippel–Lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci USA 96:12436–12441PubMedCrossRef Iwai K, Yamanaka K, Kamura T et al (1999) Identification of the von Hippel–Lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci USA 96:12436–12441PubMedCrossRef
23.
go back to reference Stebbins CE, Kaelin WG Jr, Pavletich NP (1999) Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science 284:455–461PubMedCrossRef Stebbins CE, Kaelin WG Jr, Pavletich NP (1999) Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science 284:455–461PubMedCrossRef
24.
go back to reference Mahon PC, Hirota K, Semenza GL (2001) FIH-1: a novel protein that interacts with HIF-1α and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev 15:2675–2686PubMedCrossRef Mahon PC, Hirota K, Semenza GL (2001) FIH-1: a novel protein that interacts with HIF-1α and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev 15:2675–2686PubMedCrossRef
25.
go back to reference Hewitson KS, McNeill LA, Riordan MV et al (2002) Hypoxia-inducible factor (HIF) asparagine hydroxylase is identical to factor inhibiting HIF (FIH) and is related to the cupin structural family. J Biol Chem 277:26351–26355PubMedCrossRef Hewitson KS, McNeill LA, Riordan MV et al (2002) Hypoxia-inducible factor (HIF) asparagine hydroxylase is identical to factor inhibiting HIF (FIH) and is related to the cupin structural family. J Biol Chem 277:26351–26355PubMedCrossRef
26.
go back to reference Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK (2002) FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev 16:1466–1471PubMedCrossRef Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK (2002) FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev 16:1466–1471PubMedCrossRef
27.
go back to reference Linke S, Stojkoski C, Kewley RJ, Booker GW, Whitelaw ML, Peet DJ (2004) Substrate requirements of the oxygen-sensing asparaginyl hydroxylase factor-inhibiting hypoxia-inducible factor. J Biol Chem 279:14391–14397PubMedCrossRef Linke S, Stojkoski C, Kewley RJ, Booker GW, Whitelaw ML, Peet DJ (2004) Substrate requirements of the oxygen-sensing asparaginyl hydroxylase factor-inhibiting hypoxia-inducible factor. J Biol Chem 279:14391–14397PubMedCrossRef
28.
go back to reference Safran M, Kaelin WG Jr (2003) HIF hydroxylation and the mammalian oxygen-sensing pathway. J Clin Invest 111:779–783PubMed Safran M, Kaelin WG Jr (2003) HIF hydroxylation and the mammalian oxygen-sensing pathway. J Clin Invest 111:779–783PubMed
29.
go back to reference Kimura H, Weisz A, Ogura T et al (2001) Identification of hypoxia-inducible factor 1 ancillary sequence and its function in vascular endothelial growth factor gene induction by hypoxia and nitric oxide. J Biol Chem 276:2292–2298PubMed Kimura H, Weisz A, Ogura T et al (2001) Identification of hypoxia-inducible factor 1 ancillary sequence and its function in vascular endothelial growth factor gene induction by hypoxia and nitric oxide. J Biol Chem 276:2292–2298PubMed
30.
go back to reference Carmeliet P, Dor Y, Herbert JM et al (1998) Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394:485–490PubMedCrossRef Carmeliet P, Dor Y, Herbert JM et al (1998) Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394:485–490PubMedCrossRef
31.
go back to reference Forsythe JA, Jiang BH, Iyer NV et al (1996) Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 16:4604–4613PubMed Forsythe JA, Jiang BH, Iyer NV et al (1996) Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 16:4604–4613PubMed
32.
go back to reference Hu J, Discher DJ, Bishopric NH, Webster KA (1998) Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand. Biochem Biophys Res Commun 245:894–899PubMedCrossRef Hu J, Discher DJ, Bishopric NH, Webster KA (1998) Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand. Biochem Biophys Res Commun 245:894–899PubMedCrossRef
33.
go back to reference Lee PJ, Jiang BH, Chin BY et al (1997) Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J Biol Chem 272:5375–5381PubMedCrossRef Lee PJ, Jiang BH, Chin BY et al (1997) Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J Biol Chem 272:5375–5381PubMedCrossRef
34.
go back to reference Palmer LA, Semenza GL, Stoler MH, Johns RA (1998) Hypoxia induces type II NOS gene expression in pulmonary artery endothelial cells via HIF-1. Am J Physiol 274:L212–L219PubMed Palmer LA, Semenza GL, Stoler MH, Johns RA (1998) Hypoxia induces type II NOS gene expression in pulmonary artery endothelial cells via HIF-1. Am J Physiol 274:L212–L219PubMed
35.
go back to reference Aicher A, Heeschen C, Mildner-Rihm C et al (2003) Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med 9:1370–1376PubMedCrossRef Aicher A, Heeschen C, Mildner-Rihm C et al (2003) Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med 9:1370–1376PubMedCrossRef
36.
go back to reference Thum T, Fraccarollo D, Schultheiss M et al (2007) Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes 56:666–674PubMedCrossRef Thum T, Fraccarollo D, Schultheiss M et al (2007) Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes 56:666–674PubMedCrossRef
38.
go back to reference Ceradini DJ, Kulkarni AR, Callaghan MJ et al (2004) Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10:858–864PubMedCrossRef Ceradini DJ, Kulkarni AR, Callaghan MJ et al (2004) Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10:858–864PubMedCrossRef
39.
go back to reference Zagzag D, Krishnamachary B, Yee H et al (2005) Stromal cell-derived factor-1α and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: von Hippel–Lindau loss-of-function induces expression of a ligand and its receptor. Cancer Res 65:6178–6188PubMedCrossRef Zagzag D, Krishnamachary B, Yee H et al (2005) Stromal cell-derived factor-1α and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: von Hippel–Lindau loss-of-function induces expression of a ligand and its receptor. Cancer Res 65:6178–6188PubMedCrossRef
40.
go back to reference Liu YV, Baek JH, Zhang H, Diez R, Cole RN, Semenza GL (2007) RACK1 competes with HSP90 for binding to HIF-1α and is required for O2-independent and HSP90 inhibitor-induced degradation of HIF-1α. Mol Cell 25:207–217PubMedCrossRef Liu YV, Baek JH, Zhang H, Diez R, Cole RN, Semenza GL (2007) RACK1 competes with HSP90 for binding to HIF-1α and is required for O2-independent and HSP90 inhibitor-induced degradation of HIF-1α. Mol Cell 25:207–217PubMedCrossRef
41.
go back to reference Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J (2006) Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol 26:2019–2028PubMedCrossRef Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J (2006) Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol 26:2019–2028PubMedCrossRef
42.
go back to reference Luo W, Zhong J, Chang R, Hu H, Pandey A, Semenza GL (2009) Hsp70 and CHIP selectively mediate ubiquitination and degradation of hypoxia-inducible factor (HIF)-1α but not HIF-2α. J Biol Chem 285:3651–3663PubMedCrossRef Luo W, Zhong J, Chang R, Hu H, Pandey A, Semenza GL (2009) Hsp70 and CHIP selectively mediate ubiquitination and degradation of hypoxia-inducible factor (HIF)-1α but not HIF-2α. J Biol Chem 285:3651–3663PubMedCrossRef
43.
go back to reference Bento CF, Fernandes R, Ramalho J et al (2010) The chaperone-dependent ubiquitin ligase CHIP targets HIF-1α for degradation in the presence of methylglyoxal. PLoS ONE 5:e15062PubMedCrossRef Bento CF, Fernandes R, Ramalho J et al (2010) The chaperone-dependent ubiquitin ligase CHIP targets HIF-1α for degradation in the presence of methylglyoxal. PLoS ONE 5:e15062PubMedCrossRef
44.
go back to reference Phillips SA, Thornalley PJ (1993) The formation of methylglyoxal from triose phosphates. Investigation using a specific assay for methylglyoxal. Eur J Biochem 212:101–105PubMedCrossRef Phillips SA, Thornalley PJ (1993) The formation of methylglyoxal from triose phosphates. Investigation using a specific assay for methylglyoxal. Eur J Biochem 212:101–105PubMedCrossRef
45.
go back to reference Chen D, Li M, Luo J, Gu W (2003) Direct interactions between HIF-1α and Mdm2 modulate p53 function. J Biol Chem 278:13595–13598PubMedCrossRef Chen D, Li M, Luo J, Gu W (2003) Direct interactions between HIF-1α and Mdm2 modulate p53 function. J Biol Chem 278:13595–13598PubMedCrossRef
46.
go back to reference Hansson LO, Friedler A, Freund S, Rudiger S, Fersht AR (2002) Two sequence motifs from HIF-1α bind to the DNA-binding site of p53. Proc Natl Acad Sci USA 99:10305–10309PubMedCrossRef Hansson LO, Friedler A, Freund S, Rudiger S, Fersht AR (2002) Two sequence motifs from HIF-1α bind to the DNA-binding site of p53. Proc Natl Acad Sci USA 99:10305–10309PubMedCrossRef
47.
go back to reference Tang TT, Lasky LA (2003) The forkhead transcription factor FOXO4 induces the down-regulation of hypoxia-inducible factor 1α by a von Hippel–Lindau protein-independent mechanism. J Biol Chem 278:30125–30135PubMedCrossRef Tang TT, Lasky LA (2003) The forkhead transcription factor FOXO4 induces the down-regulation of hypoxia-inducible factor 1α by a von Hippel–Lindau protein-independent mechanism. J Biol Chem 278:30125–30135PubMedCrossRef
48.
go back to reference Koh MY, Darnay BG, Powis G (2008) Hypoxia-associated factor, a novel E3-ubiquitin ligase, binds and ubiquitinates hypoxia-inducible factor 1α, leading to its oxygen-independent degradation. Mol Cell Biol 28:7081–7095PubMedCrossRef Koh MY, Darnay BG, Powis G (2008) Hypoxia-associated factor, a novel E3-ubiquitin ligase, binds and ubiquitinates hypoxia-inducible factor 1α, leading to its oxygen-independent degradation. Mol Cell Biol 28:7081–7095PubMedCrossRef
49.
go back to reference Li Z, Wang D, Messing EM, Wu G (2005) VHL protein-interacting deubiquitinating enzyme 2 deubiquitinates and stabilizes HIF-1α. EMBO Rep 6:373–378PubMedCrossRef Li Z, Wang D, Messing EM, Wu G (2005) VHL protein-interacting deubiquitinating enzyme 2 deubiquitinates and stabilizes HIF-1α. EMBO Rep 6:373–378PubMedCrossRef
50.
go back to reference Cheng J, Kang X, Zhang S, Yeh ET (2007) SUMO-specific protease 1 is essential for stabilization of HIF1α during hypoxia. Cell 131:584–595PubMedCrossRef Cheng J, Kang X, Zhang S, Yeh ET (2007) SUMO-specific protease 1 is essential for stabilization of HIF1α during hypoxia. Cell 131:584–595PubMedCrossRef
51.
go back to reference Carbia-Nagashima A, Gerez J, Perez-Castro C et al (2007) RSUME, a small RWD-containing protein, enhances SUMO conjugation and stabilizes HIF-1α during hypoxia. Cell 131:309–323PubMedCrossRef Carbia-Nagashima A, Gerez J, Perez-Castro C et al (2007) RSUME, a small RWD-containing protein, enhances SUMO conjugation and stabilizes HIF-1α during hypoxia. Cell 131:309–323PubMedCrossRef
52.
go back to reference Berta MA, Mazure N, Hattab M, Pouyssegur J, Brahimi-Horn MC (2007) SUMOylation of hypoxia-inducible factor-1α reduces its transcriptional activity. Biochem Biophys Res Commun 360:646–652PubMedCrossRef Berta MA, Mazure N, Hattab M, Pouyssegur J, Brahimi-Horn MC (2007) SUMOylation of hypoxia-inducible factor-1α reduces its transcriptional activity. Biochem Biophys Res Commun 360:646–652PubMedCrossRef
53.
go back to reference Maynard MA, Ohh M (2007) The role of hypoxia-inducible factors in cancer. Cell Mol Life Sci 64:2170–2180PubMedCrossRef Maynard MA, Ohh M (2007) The role of hypoxia-inducible factors in cancer. Cell Mol Life Sci 64:2170–2180PubMedCrossRef
54.
go back to reference Sang N, Stiehl DP, Bohensky J, Leshchinsky I, Srinivas V, Caro J (2003) MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300. J Biol Chem 278:14013–14019PubMedCrossRef Sang N, Stiehl DP, Bohensky J, Leshchinsky I, Srinivas V, Caro J (2003) MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300. J Biol Chem 278:14013–14019PubMedCrossRef
55.
go back to reference Mylonis I, Chachami G, Samiotaki M et al (2006) Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1α. J Biol Chem 281:33095–33106PubMedCrossRef Mylonis I, Chachami G, Samiotaki M et al (2006) Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1α. J Biol Chem 281:33095–33106PubMedCrossRef
56.
go back to reference Rivard A, Silver M, Chen D et al (1999) Rescue of diabetes-related impairment of angiogenesis by intramuscular gene therapy with adeno-VEGF. Am J Pathol 154:355–363PubMedCrossRef Rivard A, Silver M, Chen D et al (1999) Rescue of diabetes-related impairment of angiogenesis by intramuscular gene therapy with adeno-VEGF. Am J Pathol 154:355–363PubMedCrossRef
57.
go back to reference Sarkar K, Fox-Talbot K, Steenbergen C, Bosch-Marce M, Semenza GL (2009) Adenoviral transfer of HIF-1α enhances vascular responses to critical limb ischemia in diabetic mice. Proc Natl Acad Sci USA 106:18769–18774PubMedCrossRef Sarkar K, Fox-Talbot K, Steenbergen C, Bosch-Marce M, Semenza GL (2009) Adenoviral transfer of HIF-1α enhances vascular responses to critical limb ischemia in diabetic mice. Proc Natl Acad Sci USA 106:18769–18774PubMedCrossRef
58.
go back to reference Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA (2000) Blood-derived angioblasts accelerate blood-flow restoration in diabetic mice. J Clin Invest 106:571–578PubMedCrossRef Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA (2000) Blood-derived angioblasts accelerate blood-flow restoration in diabetic mice. J Clin Invest 106:571–578PubMedCrossRef
59.
go back to reference Botusan IR, Sunkari VG, Savu O et al (2008) Stabilization of HIF-1α is critical to improve wound healing in diabetic mice. Proc Natl Acad Sci USA 105:19426–19431PubMedCrossRef Botusan IR, Sunkari VG, Savu O et al (2008) Stabilization of HIF-1α is critical to improve wound healing in diabetic mice. Proc Natl Acad Sci USA 105:19426–19431PubMedCrossRef
60.
go back to reference Gallagher KA, Liu ZJ, Xiao M et al (2007) Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1α. J Clin Invest 117:1249–1259PubMedCrossRef Gallagher KA, Liu ZJ, Xiao M et al (2007) Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1α. J Clin Invest 117:1249–1259PubMedCrossRef
61.
go back to reference Ceradini DJ, Yao D, Grogan RH et al (2008) Decreasing intracellular superoxide corrects defective ischemia-induced new vessel formation in diabetic mice. J Biol Chem 283:10930–10938PubMedCrossRef Ceradini DJ, Yao D, Grogan RH et al (2008) Decreasing intracellular superoxide corrects defective ischemia-induced new vessel formation in diabetic mice. J Biol Chem 283:10930–10938PubMedCrossRef
62.
go back to reference Fadini GP, Sartore S, Schiavon M et al (2006) Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia–reperfusion injury in rats. Diabetologia 49:3075–3084PubMedCrossRef Fadini GP, Sartore S, Schiavon M et al (2006) Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia–reperfusion injury in rats. Diabetologia 49:3075–3084PubMedCrossRef
63.
go back to reference Liu L, Marti GP, Wei X et al (2008) Age-dependent impairment of HIF-1α expression in diabetic mice: correction with electroporation-facilitated gene therapy increases wound healing, angiogenesis, and circulating angiogenic cells. J Cell Physiol 217:319–327PubMedCrossRef Liu L, Marti GP, Wei X et al (2008) Age-dependent impairment of HIF-1α expression in diabetic mice: correction with electroporation-facilitated gene therapy increases wound healing, angiogenesis, and circulating angiogenic cells. J Cell Physiol 217:319–327PubMedCrossRef
64.
go back to reference Loomans CJ, de Koning EJ, Staal FJ et al (2004) Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes 53:195–199PubMedCrossRef Loomans CJ, de Koning EJ, Staal FJ et al (2004) Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes 53:195–199PubMedCrossRef
65.
go back to reference Tamarat R, Silvestre JS, Le Ricousse-Roussanne S et al (2004) Impairment in ischemia-induced neovascularization in diabetes: bone marrow mononuclear cell dysfunction and therapeutic potential of placenta growth factor treatment. Am J Pathol 164:457–466PubMedCrossRef Tamarat R, Silvestre JS, Le Ricousse-Roussanne S et al (2004) Impairment in ischemia-induced neovascularization in diabetes: bone marrow mononuclear cell dysfunction and therapeutic potential of placenta growth factor treatment. Am J Pathol 164:457–466PubMedCrossRef
66.
go back to reference Tepper OM, Galiano RD, Capla JM et al (2002) Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation 106:2781–2786PubMedCrossRef Tepper OM, Galiano RD, Capla JM et al (2002) Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation 106:2781–2786PubMedCrossRef
67.
go back to reference Catrina SB, Okamoto K, Pereira T, Brismar K, Poellinger L (2004) Hyperglycemia regulates hypoxia-inducible factor-1alpha protein stability and function. Diabetes 53:3226–3232PubMedCrossRef Catrina SB, Okamoto K, Pereira T, Brismar K, Poellinger L (2004) Hyperglycemia regulates hypoxia-inducible factor-1alpha protein stability and function. Diabetes 53:3226–3232PubMedCrossRef
68.
go back to reference Mace KA, Yu DH, Paydar KZ, Boudreau N, Young DM (2007) Sustained expression of Hif-1α in the diabetic environment promotes angiogenesis and cutaneous wound repair. Wound Repair Regen 15:636–645PubMedCrossRef Mace KA, Yu DH, Paydar KZ, Boudreau N, Young DM (2007) Sustained expression of Hif-1α in the diabetic environment promotes angiogenesis and cutaneous wound repair. Wound Repair Regen 15:636–645PubMedCrossRef
69.
go back to reference Chou E, Suzuma I, Way KJ et al (2002) Decreased cardiac expression of vascular endothelial growth factor and its receptors in insulin-resistant and diabetic states: a possible explanation for impaired collateral formation in cardiac tissue. Circulation 105:373–379PubMedCrossRef Chou E, Suzuma I, Way KJ et al (2002) Decreased cardiac expression of vascular endothelial growth factor and its receptors in insulin-resistant and diabetic states: a possible explanation for impaired collateral formation in cardiac tissue. Circulation 105:373–379PubMedCrossRef
70.
go back to reference Baelde HJ, Eikmans M, Lappin DW et al (2007) Reduction of VEGF-A and CTGF expression in diabetic nephropathy is associated with podocyte loss. Kidney Int 71:637–645PubMedCrossRef Baelde HJ, Eikmans M, Lappin DW et al (2007) Reduction of VEGF-A and CTGF expression in diabetic nephropathy is associated with podocyte loss. Kidney Int 71:637–645PubMedCrossRef
71.
go back to reference Wirostko B, Wong TY, Simo R (2008) Vascular endothelial growth factor and diabetic complications. Prog Retin Eye Res 27:608–621PubMedCrossRef Wirostko B, Wong TY, Simo R (2008) Vascular endothelial growth factor and diabetic complications. Prog Retin Eye Res 27:608–621PubMedCrossRef
72.
go back to reference Duh E, Aiello LP (1999) Vascular endothelial growth factor and diabetes: the agonist vs antagonist paradox. Diabetes 48:1899–1906PubMedCrossRef Duh E, Aiello LP (1999) Vascular endothelial growth factor and diabetes: the agonist vs antagonist paradox. Diabetes 48:1899–1906PubMedCrossRef
73.
go back to reference Yoon YS, Uchida S, Masuo O et al (2005) Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy: restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor. Circulation 111:2073–2085PubMedCrossRef Yoon YS, Uchida S, Masuo O et al (2005) Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy: restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor. Circulation 111:2073–2085PubMedCrossRef
74.
go back to reference Price SA, Dent C, Duran-Jimenez B et al (2006) Gene transfer of an engineered transcription factor promoting expression of VEGF-A protects against experimental diabetic neuropathy. Diabetes 55:1847–1854PubMedCrossRef Price SA, Dent C, Duran-Jimenez B et al (2006) Gene transfer of an engineered transcription factor promoting expression of VEGF-A protects against experimental diabetic neuropathy. Diabetes 55:1847–1854PubMedCrossRef
75.
go back to reference Murakami T, Arai M, Sunada Y, Nakamura A (2006) VEGF 164 gene transfer by electroporation improves diabetic sensory neuropathy in mice. J Gene Med 8:773–781PubMedCrossRef Murakami T, Arai M, Sunada Y, Nakamura A (2006) VEGF 164 gene transfer by electroporation improves diabetic sensory neuropathy in mice. J Gene Med 8:773–781PubMedCrossRef
76.
go back to reference Chattopadhyay M, Krisky D, Wolfe D, Glorioso JC, Mata M, Fink DJ (2005) HSV-mediated gene transfer of vascular endothelial growth factor to dorsal root ganglia prevents diabetic neuropathy. Gene Ther 12:1377–1384PubMedCrossRef Chattopadhyay M, Krisky D, Wolfe D, Glorioso JC, Mata M, Fink DJ (2005) HSV-mediated gene transfer of vascular endothelial growth factor to dorsal root ganglia prevents diabetic neuropathy. Gene Ther 12:1377–1384PubMedCrossRef
77.
go back to reference Thangarajah H, Yao D, Chang EI et al (2009) The molecular basis for impaired hypoxia-induced VEGF expression in diabetic tissues. Proc Natl Acad Sci USA 106:13505–13510PubMedCrossRef Thangarajah H, Yao D, Chang EI et al (2009) The molecular basis for impaired hypoxia-induced VEGF expression in diabetic tissues. Proc Natl Acad Sci USA 106:13505–13510PubMedCrossRef
78.
go back to reference Malmberg K, Norhammar A, Wedel H, Ryden L (1999) Glycometabolic state at admission: important risk marker of mortality in conventionally treated patients with diabetes mellitus and acute myocardial infarction: long-term results from the Diabetes and Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) study. Circulation 99:2626–2632PubMed Malmberg K, Norhammar A, Wedel H, Ryden L (1999) Glycometabolic state at admission: important risk marker of mortality in conventionally treated patients with diabetes mellitus and acute myocardial infarction: long-term results from the Diabetes and Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) study. Circulation 99:2626–2632PubMed
79.
go back to reference Teixeira AS, Andrade SP (1999) Glucose-induced inhibition of angiogenesis in the rat sponge granuloma is prevented by aminoguanidine. Life Sci 64:655–662PubMedCrossRef Teixeira AS, Andrade SP (1999) Glucose-induced inhibition of angiogenesis in the rat sponge granuloma is prevented by aminoguanidine. Life Sci 64:655–662PubMedCrossRef
80.
go back to reference Tamarat R, Silvestre JS, Huijberts M et al (2003) Blockade of advanced glycation end-product formation restores ischemia-induced angiogenesis in diabetic mice. Proc Natl Acad Sci USA 100:8555–8560PubMedCrossRef Tamarat R, Silvestre JS, Huijberts M et al (2003) Blockade of advanced glycation end-product formation restores ischemia-induced angiogenesis in diabetic mice. Proc Natl Acad Sci USA 100:8555–8560PubMedCrossRef
81.
go back to reference Bento CF, Fernandes R, Matafome P, Sena C, Seica R, Pereira P (2010) Methylglyoxal-induced imbalance in the ratio of vascular endothelial growth factor to angiopoietin 2 secreted by retinal pigment epithelial cells leads to endothelial dysfunction. Exp Physiol 95:955–970PubMed Bento CF, Fernandes R, Matafome P, Sena C, Seica R, Pereira P (2010) Methylglyoxal-induced imbalance in the ratio of vascular endothelial growth factor to angiopoietin 2 secreted by retinal pigment epithelial cells leads to endothelial dysfunction. Exp Physiol 95:955–970PubMed
82.
go back to reference Abaci A, Oguzhan A, Kahraman S et al (1999) Effect of diabetes mellitus on formation of coronary collateral vessels. Circulation 99:2239–2242PubMed Abaci A, Oguzhan A, Kahraman S et al (1999) Effect of diabetes mellitus on formation of coronary collateral vessels. Circulation 99:2239–2242PubMed
83.
go back to reference Larger E, Marre M, Corvol P, Gasc JM (2004) Hyperglycemia-induced defects in angiogenesis in the chicken chorioallantoic membrane model. Diabetes 53:752–761PubMedCrossRef Larger E, Marre M, Corvol P, Gasc JM (2004) Hyperglycemia-induced defects in angiogenesis in the chicken chorioallantoic membrane model. Diabetes 53:752–761PubMedCrossRef
84.
go back to reference Marfella R, D’Amico M, Di Filippo C et al (2002) Myocardial infarction in diabetic rats: role of hyperglycaemia on infarct size and early expression of hypoxia-inducible factor 1. Diabetologia 45:1172–1181PubMedCrossRef Marfella R, D’Amico M, Di Filippo C et al (2002) Myocardial infarction in diabetic rats: role of hyperglycaemia on infarct size and early expression of hypoxia-inducible factor 1. Diabetologia 45:1172–1181PubMedCrossRef
85.
go back to reference Xue W, Cai L, Tan Y et al (2010) Cardiac-specific overexpression of HIF-1α prevents deterioration of glycolytic pathway and cardiac remodeling in streptozotocin-induced diabetic mice. Am J Pathol 177:97–105PubMedCrossRef Xue W, Cai L, Tan Y et al (2010) Cardiac-specific overexpression of HIF-1α prevents deterioration of glycolytic pathway and cardiac remodeling in streptozotocin-induced diabetic mice. Am J Pathol 177:97–105PubMedCrossRef
86.
go back to reference Gunton JE, Kulkarni RN, Yim S et al (2005) Loss of ARNT/HIF1β mediates altered gene expression and pancreatic-islet dysfunction in human type 2 diabetes. Cell 122:337–349PubMedCrossRef Gunton JE, Kulkarni RN, Yim S et al (2005) Loss of ARNT/HIF1β mediates altered gene expression and pancreatic-islet dysfunction in human type 2 diabetes. Cell 122:337–349PubMedCrossRef
87.
go back to reference Nagy G, Kovacs-Nagy R, Kereszturi E et al (2009) Association of hypoxia inducible factor-1 α gene polymorphism with both type 1 and type 2 diabetes in a Caucasian (Hungarian) sample. BMC Med Genet 10:79PubMedCrossRef Nagy G, Kovacs-Nagy R, Kereszturi E et al (2009) Association of hypoxia inducible factor-1 α gene polymorphism with both type 1 and type 2 diabetes in a Caucasian (Hungarian) sample. BMC Med Genet 10:79PubMedCrossRef
88.
go back to reference Yamada N, Horikawa Y, Oda N et al (2005) Genetic variation in the hypoxia-inducible factor-1α gene is associated with type 2 diabetes in Japanese. J Clin Endocrinol Metab 90:5841–5847PubMedCrossRef Yamada N, Horikawa Y, Oda N et al (2005) Genetic variation in the hypoxia-inducible factor-1α gene is associated with type 2 diabetes in Japanese. J Clin Endocrinol Metab 90:5841–5847PubMedCrossRef
89.
go back to reference Resar JR, Roguin A, Voner J et al (2005) Hypoxia-inducible factor 1α polymorphism and coronary collaterals in patients with ischemic heart disease. Chest 128:787–791PubMedCrossRef Resar JR, Roguin A, Voner J et al (2005) Hypoxia-inducible factor 1α polymorphism and coronary collaterals in patients with ischemic heart disease. Chest 128:787–791PubMedCrossRef
90.
go back to reference Isoe T, Makino Y, Mizumoto K et al (2010) High glucose activates HIF-1-mediated signal transduction in glomerular mesangial cells through a carbohydrate response element binding protein. Kidney Int 78:48–59PubMedCrossRef Isoe T, Makino Y, Mizumoto K et al (2010) High glucose activates HIF-1-mediated signal transduction in glomerular mesangial cells through a carbohydrate response element binding protein. Kidney Int 78:48–59PubMedCrossRef
91.
go back to reference Makino H, Miyamoto Y, Sawai K et al (2006) Altered gene expression related to glomerulogenesis and podocyte structure in early diabetic nephropathy of db/db mice and its restoration by pioglitazone. Diabetes 55:2747–2756PubMedCrossRef Makino H, Miyamoto Y, Sawai K et al (2006) Altered gene expression related to glomerulogenesis and podocyte structure in early diabetic nephropathy of db/db mice and its restoration by pioglitazone. Diabetes 55:2747–2756PubMedCrossRef
92.
go back to reference Guo S, Bragina O, Xu Y et al (2008) Glucose up-regulates HIF-1α expression in primary cortical neurons in response to hypoxia through maintaining cellular redox status. J Neurochem 105:1849–1860PubMedCrossRef Guo S, Bragina O, Xu Y et al (2008) Glucose up-regulates HIF-1α expression in primary cortical neurons in response to hypoxia through maintaining cellular redox status. J Neurochem 105:1849–1860PubMedCrossRef
93.
go back to reference Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van Obberghen E (2002) Insulin stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapamycin-dependent signaling pathway. J Biol Chem 277:27975–27981PubMedCrossRef Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van Obberghen E (2002) Insulin stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapamycin-dependent signaling pathway. J Biol Chem 277:27975–27981PubMedCrossRef
94.
go back to reference Thornalley PJ (2003) Glyoxalase I—structure, function and a critical role in the enzymatic defence against glycation. Biochem Soc Trans 31:1343–1348PubMedCrossRef Thornalley PJ (2003) Glyoxalase I—structure, function and a critical role in the enzymatic defence against glycation. Biochem Soc Trans 31:1343–1348PubMedCrossRef
95.
go back to reference Dinçer Y, Alademir Z, Ilkova H, Akçay T (2002) Susceptibility of glutatione and glutathione-related antioxidant activity to hydrogen peroxide in patients with type 2 diabetes: effect of glycemic control. Clin Biochem 35:297–301PubMedCrossRef Dinçer Y, Alademir Z, Ilkova H, Akçay T (2002) Susceptibility of glutatione and glutathione-related antioxidant activity to hydrogen peroxide in patients with type 2 diabetes: effect of glycemic control. Clin Biochem 35:297–301PubMedCrossRef
96.
go back to reference Poulaki V, Qin W, Joussen AM et al (2002) Acute intensive insulin therapy exacerbates diabetic blood–retinal barrier breakdown via hypoxia-inducible factor-1α and VEGF. J Clin Invest 109:805–815PubMed Poulaki V, Qin W, Joussen AM et al (2002) Acute intensive insulin therapy exacerbates diabetic blood–retinal barrier breakdown via hypoxia-inducible factor-1α and VEGF. J Clin Invest 109:805–815PubMed
Metadata
Title
Regulation of hypoxia-inducible factor 1 and the loss of the cellular response to hypoxia in diabetes
Authors
C. F. Bento
P. Pereira
Publication date
01-08-2011
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 8/2011
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-011-2191-8

Other articles of this Issue 8/2011

Diabetologia 8/2011 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.