Skip to main content
Top
Published in: Journal of Inherited Metabolic Disease 4/2017

01-07-2017 | SSIEM 2016

Inherited disorders of transition metal metabolism: an update

Author: Peter T. Clayton

Published in: Journal of Inherited Metabolic Disease | Issue 4/2017

Login to get access

Abstract

Elements with a biological role include six trace transition metals: manganese, iron, cobalt, copper, zinc and molybdenum. Transition metals participate in group transfer reactions such as glycosylation and phosphorylation and those that can transfer an electron by alternating between two redox states such as iron (3+/2+) and copper (2+/1+) are also very important in biological redox reactions including the reduction of molecular oxygen and the transport of oxygen. However, these trace metals are also potentially toxic, generating reactive oxygen species through Fenton chemistry. Recently, a role of trace metals in host defence (“nutritional immunity”) has been recognized. The host can deprive the pathogen of a trace metal or poison it with a toxic concentration. Disorders leading to low concentrations of a trace metal can often be treated by supplementing that metal; disorders leading to excessively high concentrations can often be treated with chelating agents such as penicillamine and disodium calcium edetate. This update will address: i) the manganese/zinc transporters (because two new treatable disorders were described in 2016 – SLC39A8 deficiency and SLC39A14 deficiency); ii) copper transporter disorders because we need to improve the treatment of patients with neurological symptoms due to Wilson’s disease; and iii) iron homeostasis because recent progress in research into the metabolism of iron and its regulation helps us better understand several inborn errors affecting these pathways.
Literature
go back to reference Ahting U, Mayr JA, Vanlander AV, Hardy SA, Santra S, Makowski C, Alston CL, Zimmermann FA, Abela L, Plecko B, Rohrbach M, Spranger S, Seneca S, Rolinski B, Hagendorff A, Hempel M, Sperl W, Meitinger T, Smet J, Taylor RW, Van Coster R, Freisinger P, Prokisch H, Haack TB (2015) Clinical, biochemical, and genetic spectrum of seven patients with NFU1 deficiency. Front Genet 6:123CrossRefPubMedPubMedCentral Ahting U, Mayr JA, Vanlander AV, Hardy SA, Santra S, Makowski C, Alston CL, Zimmermann FA, Abela L, Plecko B, Rohrbach M, Spranger S, Seneca S, Rolinski B, Hagendorff A, Hempel M, Sperl W, Meitinger T, Smet J, Taylor RW, Van Coster R, Freisinger P, Prokisch H, Haack TB (2015) Clinical, biochemical, and genetic spectrum of seven patients with NFU1 deficiency. Front Genet 6:123CrossRefPubMedPubMedCentral
go back to reference Barupala DP, Dzul SP, Riggs-Gelasco PJ, Stemmler TL (2016) Synthesis, delivery and regulation of eukaryotic heme and Fe-S cluster cofactors. Arch Biochem Biophys 592:60–75CrossRefPubMedPubMedCentral Barupala DP, Dzul SP, Riggs-Gelasco PJ, Stemmler TL (2016) Synthesis, delivery and regulation of eukaryotic heme and Fe-S cluster cofactors. Arch Biochem Biophys 592:60–75CrossRefPubMedPubMedCentral
go back to reference Boycott KM, Beaulieu CL, Kernohan KD, Gebril OH, Mhanni A, Chudley AE, Redl D, Qin W, Hampson S, Küry S, Tetreault M, Puffenberger EG, Scott JN, Bezieau S, Reis A, Uebe S, Schumacher J, Hegele RA, McLeod DR, Gálvez-Peralta M, Majewski J, Ramaekers VT, Care4Rare Canada Consortium, Nebert DW, Innes AM, Parboosingh JS, Abou Jamra R (2015) Autosomal-recessive intellectual disability with cerebellar atrophy syndrome caused by mutation of the manganese and zinc transporter gene SLC39A8. Am J Hum Genet 97(6):886–893CrossRefPubMedPubMedCentral Boycott KM, Beaulieu CL, Kernohan KD, Gebril OH, Mhanni A, Chudley AE, Redl D, Qin W, Hampson S, Küry S, Tetreault M, Puffenberger EG, Scott JN, Bezieau S, Reis A, Uebe S, Schumacher J, Hegele RA, McLeod DR, Gálvez-Peralta M, Majewski J, Ramaekers VT, Care4Rare Canada Consortium, Nebert DW, Innes AM, Parboosingh JS, Abou Jamra R (2015) Autosomal-recessive intellectual disability with cerebellar atrophy syndrome caused by mutation of the manganese and zinc transporter gene SLC39A8. Am J Hum Genet 97(6):886–893CrossRefPubMedPubMedCentral
go back to reference Canonne-Hergaux F, Gruenheid S, Govoni G, Gros P (1999) The Nramp1 protein and its role in resistance to infection and macrophage function. Proc Assoc Am Physicians 111(4):283–289CrossRefPubMed Canonne-Hergaux F, Gruenheid S, Govoni G, Gros P (1999) The Nramp1 protein and its role in resistance to infection and macrophage function. Proc Assoc Am Physicians 111(4):283–289CrossRefPubMed
go back to reference Chen P, Chakraborty S, Mukhopadhyay S, Lee E, Paoliello MM, Bowman AB, Aschner M (2015) Manganese homeostasis in the nervous system. J Neurochem 134(4):601–610CrossRefPubMedPubMedCentral Chen P, Chakraborty S, Mukhopadhyay S, Lee E, Paoliello MM, Bowman AB, Aschner M (2015) Manganese homeostasis in the nervous system. J Neurochem 134(4):601–610CrossRefPubMedPubMedCentral
go back to reference Chesi G, Hegde RN, Iacobacci S, Concilli M, Parashuraman S, Festa BP, Polishchuk EV, Di Tullio G, Carissimo A, Montefusco S, Canetti D, Monti M, Amoresano A, Pucci P, van de Sluis B, Lutsenko S, Luini A, Polishchuk RS (2016) Identification of p38 MAPK and JNK as new targets for correction of Wilson disease-causing ATP7B mutants. Hepatology 63(6):1842–1859CrossRefPubMedPubMedCentral Chesi G, Hegde RN, Iacobacci S, Concilli M, Parashuraman S, Festa BP, Polishchuk EV, Di Tullio G, Carissimo A, Montefusco S, Canetti D, Monti M, Amoresano A, Pucci P, van de Sluis B, Lutsenko S, Luini A, Polishchuk RS (2016) Identification of p38 MAPK and JNK as new targets for correction of Wilson disease-causing ATP7B mutants. Hepatology 63(6):1842–1859CrossRefPubMedPubMedCentral
go back to reference Chowanadisai W, Lönnerdal B, Kelleher SL (2006) Identification of a mutation in SLC30A2 (ZnT-2) in women with low milk zinc concentration that results in transient neonatal zinc deficiency. J Biol Chem 281(51):39699–39707CrossRefPubMed Chowanadisai W, Lönnerdal B, Kelleher SL (2006) Identification of a mutation in SLC30A2 (ZnT-2) in women with low milk zinc concentration that results in transient neonatal zinc deficiency. J Biol Chem 281(51):39699–39707CrossRefPubMed
go back to reference Członkowska A, Litwin T, Karliński M, Dziezyc K, Chabik G, Czerska M (2014) D-penicillamine versus zinc sulfate as first-line therapy for Wilson’s disease. Eur J Neurol 21(4):599–606CrossRefPubMed Członkowska A, Litwin T, Karliński M, Dziezyc K, Chabik G, Czerska M (2014) D-penicillamine versus zinc sulfate as first-line therapy for Wilson’s disease. Eur J Neurol 21(4):599–606CrossRefPubMed
go back to reference Dhawan A, Taylor RM, Cheeseman P, De Silva P, Katsiyiannakis L, Mieli-Vergani G (2005) Wilson’s disease in children: 37-year experience and revised King’s score for liver transplantation. Liver Transpl 11(4):441–448CrossRefPubMed Dhawan A, Taylor RM, Cheeseman P, De Silva P, Katsiyiannakis L, Mieli-Vergani G (2005) Wilson’s disease in children: 37-year experience and revised King’s score for liver transplantation. Liver Transpl 11(4):441–448CrossRefPubMed
go back to reference Garringer HJ, Irimia JM, Li W, Goodwin CB, Richine B, Acton A, Chan RJ, Peacock M, Muhoberac BB, Ghetti B, Vidal R (2016) Effect of systemic iron overload and a chelation therapy in a mouse model of the neurodegenerative disease hereditary ferritinopathy. PLoS One 11(8):e0161341CrossRefPubMedPubMedCentral Garringer HJ, Irimia JM, Li W, Goodwin CB, Richine B, Acton A, Chan RJ, Peacock M, Muhoberac BB, Ghetti B, Vidal R (2016) Effect of systemic iron overload and a chelation therapy in a mouse model of the neurodegenerative disease hereditary ferritinopathy. PLoS One 11(8):e0161341CrossRefPubMedPubMedCentral
go back to reference Kambe T, Tsuji T, Hashimoto A, Itsumura N (2015) The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 95(3):749–784CrossRefPubMed Kambe T, Tsuji T, Hashimoto A, Itsumura N (2015) The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 95(3):749–784CrossRefPubMed
go back to reference Küry S, Dréno B, Bézieau S, Giraudet S, Kharfi M, Kamoun R, Moisan JP (2002) Identification of SLC39A4, a gene involved in acrodermatitis enteropathica. Nat Genet 31(3):239–240CrossRefPubMed Küry S, Dréno B, Bézieau S, Giraudet S, Kharfi M, Kamoun R, Moisan JP (2002) Identification of SLC39A4, a gene involved in acrodermatitis enteropathica. Nat Genet 31(3):239–240CrossRefPubMed
go back to reference Lichtmannegger J, Leitzinger C, Wimmer R, Schmitt S, Schulz S, Kabiri Y, Eberhagen C, Rieder T, Janik D, Neff F, Straub BK, Schirmacher P, DiSpirito AA, Bandow N, Baral BS, Flatley A, Kremmer E, Denk G, Reiter FP, Hohenester S, Eckardt-Schupp F, Dencher NA, Adamski J, Sauer V, Niemietz C, Schmidt HH, Merle U, Gotthardt DN, Kroemer G, Weiss KH, Zischka H (2016) Methanobactin reverses acute liver failure in a rat model of Wilson disease. J Clin Invest 126(7):2721–2735CrossRefPubMedPubMedCentral Lichtmannegger J, Leitzinger C, Wimmer R, Schmitt S, Schulz S, Kabiri Y, Eberhagen C, Rieder T, Janik D, Neff F, Straub BK, Schirmacher P, DiSpirito AA, Bandow N, Baral BS, Flatley A, Kremmer E, Denk G, Reiter FP, Hohenester S, Eckardt-Schupp F, Dencher NA, Adamski J, Sauer V, Niemietz C, Schmidt HH, Merle U, Gotthardt DN, Kroemer G, Weiss KH, Zischka H (2016) Methanobactin reverses acute liver failure in a rat model of Wilson disease. J Clin Invest 126(7):2721–2735CrossRefPubMedPubMedCentral
go back to reference Litwin T, Dzieżyc K, Karliński M, Chabik G, Czepiel W, Członkowska A (2015) Early neurological worsening in patients with Wilson’s disease. J Neurol Sci 355(1-2):162–167CrossRefPubMed Litwin T, Dzieżyc K, Karliński M, Chabik G, Czepiel W, Członkowska A (2015) Early neurological worsening in patients with Wilson’s disease. J Neurol Sci 355(1-2):162–167CrossRefPubMed
go back to reference Martinelli D, Dionisi-Vici C (2014) AP1S1 defect causing MEDNIK syndrome: a new adaptinopathy associated with defective copper metabolism. Ann N Y Acad Sci 1314:55–63CrossRefPubMed Martinelli D, Dionisi-Vici C (2014) AP1S1 defect causing MEDNIK syndrome: a new adaptinopathy associated with defective copper metabolism. Ann N Y Acad Sci 1314:55–63CrossRefPubMed
go back to reference Martinelli D, Travaglini L, Drouin CA, Ceballos-Picot I, Rizza T, Bertini E, Carrozzo R, Petrini S, de Lonlay P, El Hachem M, Hubert L, Montpetit A, Torre G, Dionisi-Vici C (2013) MEDNIK syndrome: a novel defect of copper metabolism treatable by zinc acetate therapy. Brain 136:872–881CrossRefPubMed Martinelli D, Travaglini L, Drouin CA, Ceballos-Picot I, Rizza T, Bertini E, Carrozzo R, Petrini S, de Lonlay P, El Hachem M, Hubert L, Montpetit A, Torre G, Dionisi-Vici C (2013) MEDNIK syndrome: a novel defect of copper metabolism treatable by zinc acetate therapy. Brain 136:872–881CrossRefPubMed
go back to reference Miyajima H, Takahashi Y, Kamata T, Shimizu H, Sakai N, Gitlin JD (1997) Use of desferrioxamine in the treatment of aceruloplasminemia. Ann Neurol 41(3):404–407CrossRefPubMed Miyajima H, Takahashi Y, Kamata T, Shimizu H, Sakai N, Gitlin JD (1997) Use of desferrioxamine in the treatment of aceruloplasminemia. Ann Neurol 41(3):404–407CrossRefPubMed
go back to reference Moalem S, Percy ME, Andrews DF, Kruck TP, Wong S, Dalton AJ, Mehta P, Fedor B, Warren AC (2000) Are hereditary hemochromatosis mutations involved in Alzheimer disease? Am J Med Genet 93(1):58–66CrossRefPubMed Moalem S, Percy ME, Andrews DF, Kruck TP, Wong S, Dalton AJ, Mehta P, Fedor B, Warren AC (2000) Are hereditary hemochromatosis mutations involved in Alzheimer disease? Am J Med Genet 93(1):58–66CrossRefPubMed
go back to reference Niederau C1, Fischer R, Pürschel A, Stremmel W, Häussinger D, Strohmeyer G. (1996) Long-term survival in patients with hereditary hemochromatosis. Gastroenterology 110(4):1107--19 Niederau C1, Fischer R, Pürschel A, Stremmel W, Häussinger D, Strohmeyer G. (1996) Long-term survival in patients with hereditary hemochromatosis. Gastroenterology 110(4):1107--19
go back to reference Park JH, Hogrebe M, Grüneberg M, DuChesne I, von der Heiden AL, Reunert J, Schlingmann KP, Boycott KM, Beaulieu CL, Mhanni AA, Innes AM, Hörtnagel K, Biskup S, Gleixner EM, Kurlemann G, Fiedler B, Omran H, Rutsch F, Wada Y, Tsiakas K, Santer R, Nebert DW, Rust S, Marquardt T (2015) SLC39A8 Deficiency: A Disorder of manganese transport and glycosylation. Am J Hum Genet 97(6):894--903 Park JH, Hogrebe M, Grüneberg M, DuChesne I, von der Heiden AL, Reunert J, Schlingmann KP, Boycott KM, Beaulieu CL, Mhanni AA, Innes AM, Hörtnagel K, Biskup S, Gleixner EM, Kurlemann G, Fiedler B, Omran H, Rutsch F, Wada Y, Tsiakas K, Santer R, Nebert DW, Rust S, Marquardt T (2015) SLC39A8 Deficiency: A Disorder of manganese transport and glycosylation. Am J Hum Genet 97(6):894--903
go back to reference Park J H, Hogrebe M, Grueneberg M, Reunert J, Rust S, Marquardt T (2016) SLC39A8 deficiency is a novel treatable disorder of manganese metabolism and glycosylation. J Inherit Metab Dis 39(Issue 1 Supplement S54) Park J H, Hogrebe M, Grueneberg M, Reunert J, Rust S, Marquardt T (2016) SLC39A8 deficiency is a novel treatable disorder of manganese metabolism and glycosylation. J Inherit Metab Dis 39(Issue 1 Supplement S54)
go back to reference Quadri M, Federico A, Zhao T, Breedveld GJ, Battisti C, Delnooz C, Severijnen LA, Di Toro Mammarella L, Mignarri A, Monti L, Sanna A, Lu P, Punzo F, Cossu G, Willemsen R, Rasi F, Oostra BA, van de Warrenburg BP, Bonifati V (2012) Mutations in SLC30A10 cause parkinsonism and dystonia with hypermanganesemia, polycythemia, and chronic liver disease. Am J Hum Genet 90(3):467–477CrossRefPubMedPubMedCentral Quadri M, Federico A, Zhao T, Breedveld GJ, Battisti C, Delnooz C, Severijnen LA, Di Toro Mammarella L, Mignarri A, Monti L, Sanna A, Lu P, Punzo F, Cossu G, Willemsen R, Rasi F, Oostra BA, van de Warrenburg BP, Bonifati V (2012) Mutations in SLC30A10 cause parkinsonism and dystonia with hypermanganesemia, polycythemia, and chronic liver disease. Am J Hum Genet 90(3):467–477CrossRefPubMedPubMedCentral
go back to reference Rahman S, Mayr JA (2016) Disorders of oxidative phosphorylation. In: Saudubray JM et al (eds) Inborn metabolic diseases. Springer, Berlin Rahman S, Mayr JA (2016) Disorders of oxidative phosphorylation. In: Saudubray JM et al (eds) Inborn metabolic diseases. Springer, Berlin
go back to reference Ramakrishnan B, Ramasamy V, Qasba PK (2006) Structural snapshots of beta-1,4-galactosyltransferase-I along the kinetic pathway.J. Mol Biol 357(5):1619–1633CrossRef Ramakrishnan B, Ramasamy V, Qasba PK (2006) Structural snapshots of beta-1,4-galactosyltransferase-I along the kinetic pathway.J. Mol Biol 357(5):1619–1633CrossRef
go back to reference Riley LG, Cowley MJ, Gayevskiy V, Roscioli T, Thorburn DR, Prelog K, Bahlo M, Sue CM, Balasubramaniam S, Christodoulou J (2016) A SLC39A8 variant causes manganese deficiency, and glycosylation and mitochondrial disorders. J Inherit Metab Dis. doi:10.1007/s10545-016-0010-6 Riley LG, Cowley MJ, Gayevskiy V, Roscioli T, Thorburn DR, Prelog K, Bahlo M, Sue CM, Balasubramaniam S, Christodoulou J (2016) A SLC39A8 variant causes manganese deficiency, and glycosylation and mitochondrial disorders. J Inherit Metab Dis. doi:10.1007/s10545-016-0010-6
go back to reference Schlaug G, Hefter H, Engelbrecht V, Kuwert T, Arnold S, Stöcklin G, Seitz RJ (1996) Neurological impairment and recovery in Wislon’s disease: evidence from PET and MRI. J Neurol Sci 136:129–139CrossRefPubMed Schlaug G, Hefter H, Engelbrecht V, Kuwert T, Arnold S, Stöcklin G, Seitz RJ (1996) Neurological impairment and recovery in Wislon’s disease: evidence from PET and MRI. J Neurol Sci 136:129–139CrossRefPubMed
go back to reference Sharma V, Karlin KD, Wikström M (2013) Computational study of the activated O(H) state in the catalytic mechanism of cytochrome c oxidase. Proc Natl Acad Sci U S A 110(42):16844–16849CrossRefPubMedPubMedCentral Sharma V, Karlin KD, Wikström M (2013) Computational study of the activated O(H) state in the catalytic mechanism of cytochrome c oxidase. Proc Natl Acad Sci U S A 110(42):16844–16849CrossRefPubMedPubMedCentral
go back to reference Tuschl K, Mills PB, Parsons H, Malone M, Fowler D, Bitner-Glindzicz M, Clayton PT (2008) Hepatic cirrhosis, dystonia, polycythaemia and hypermanganesaemia--a new metabolic disorder. J Inherit Metab Dis 31(2):151–163CrossRefPubMed Tuschl K, Mills PB, Parsons H, Malone M, Fowler D, Bitner-Glindzicz M, Clayton PT (2008) Hepatic cirrhosis, dystonia, polycythaemia and hypermanganesaemia--a new metabolic disorder. J Inherit Metab Dis 31(2):151–163CrossRefPubMed
go back to reference Tuschl K, Clayton PT, Gospe SM Jr, Gulab S, Ibrahim S, Singhi P, Aulakh R, Ribeiro RT, Barsottini OG, Zaki MS, Del Rosario ML, Dyack S, Price V, Rideout A, Gordon K, Wevers RA, Chong WK, Mills PB (2012) Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man. Am J Hum Genet 90(3):457–466CrossRefPubMedPubMedCentral Tuschl K, Clayton PT, Gospe SM Jr, Gulab S, Ibrahim S, Singhi P, Aulakh R, Ribeiro RT, Barsottini OG, Zaki MS, Del Rosario ML, Dyack S, Price V, Rideout A, Gordon K, Wevers RA, Chong WK, Mills PB (2012) Syndrome of hepatic cirrhosis, dystonia, polycythemia, and hypermanganesemia caused by mutations in SLC30A10, a manganese transporter in man. Am J Hum Genet 90(3):457–466CrossRefPubMedPubMedCentral
go back to reference Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, Hung CY, Simpson MA, Chong WK, Jacques TS, Woltjer RL, Eaton S, Gregory A, Sanford L, Kara E, Houlden H, Cuno SM, Prokisch H, Valletta L, Tiranti V, Younis R, Maher ER, Spencer J, Straatman-Iwanowska A, Gissen P, Selim LA, Pintos-Morell G, Coroleu-Lletget W, Mohammad SS, Yoganathan S, Dale RC, Thomas M, Rihel J, Bodamer OA, Enns CA, Hayflick SJ, Clayton PT, Mills PB, Kurian MA, Wilson SW (2016) Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun 7:11601CrossRefPubMedPubMedCentral Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, Hung CY, Simpson MA, Chong WK, Jacques TS, Woltjer RL, Eaton S, Gregory A, Sanford L, Kara E, Houlden H, Cuno SM, Prokisch H, Valletta L, Tiranti V, Younis R, Maher ER, Spencer J, Straatman-Iwanowska A, Gissen P, Selim LA, Pintos-Morell G, Coroleu-Lletget W, Mohammad SS, Yoganathan S, Dale RC, Thomas M, Rihel J, Bodamer OA, Enns CA, Hayflick SJ, Clayton PT, Mills PB, Kurian MA, Wilson SW (2016) Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun 7:11601CrossRefPubMedPubMedCentral
go back to reference van Hasselt PM, Clayton P, Houwen RHJ (2016) Disorders in the transport of copper, iron, magnesium, manganese, selenium and zinc. In: Saudubray JM, Baumgartner MR, Walter J (eds) Inborn metabolic diseases. Diagnosis and treatment, 6th edn. Springer, Berlin van Hasselt PM, Clayton P, Houwen RHJ (2016) Disorders in the transport of copper, iron, magnesium, manganese, selenium and zinc. In: Saudubray JM, Baumgartner MR, Walter J (eds) Inborn metabolic diseases. Diagnosis and treatment, 6th edn. Springer, Berlin
go back to reference Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR (2004) Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci 5(11):863–873CrossRefPubMed Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR (2004) Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci 5(11):863–873CrossRefPubMed
Metadata
Title
Inherited disorders of transition metal metabolism: an update
Author
Peter T. Clayton
Publication date
01-07-2017
Publisher
Springer Netherlands
Published in
Journal of Inherited Metabolic Disease / Issue 4/2017
Print ISSN: 0141-8955
Electronic ISSN: 1573-2665
DOI
https://doi.org/10.1007/s10545-017-0030-x

Other articles of this Issue 4/2017

Journal of Inherited Metabolic Disease 4/2017 Go to the issue

Highlights

News and views

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.